WO2012106385A2 - Methods of identifying multiple epitopes in cells - Google Patents

Methods of identifying multiple epitopes in cells Download PDF

Info

Publication number
WO2012106385A2
WO2012106385A2 PCT/US2012/023411 US2012023411W WO2012106385A2 WO 2012106385 A2 WO2012106385 A2 WO 2012106385A2 US 2012023411 W US2012023411 W US 2012023411W WO 2012106385 A2 WO2012106385 A2 WO 2012106385A2
Authority
WO
WIPO (PCT)
Prior art keywords
esb
aps
uba
apss
target molecule
Prior art date
Application number
PCT/US2012/023411
Other languages
English (en)
French (fr)
Other versions
WO2012106385A3 (en
Inventor
Gary P. NOLAN
Original Assignee
Apprise Bio, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=46603278&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2012106385(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Apprise Bio, Inc. filed Critical Apprise Bio, Inc.
Priority to JP2013551435A priority Critical patent/JP6069224B2/ja
Priority to CN201280016828.5A priority patent/CN103703143B/zh
Priority to US13/981,711 priority patent/US10144950B2/en
Priority to EP12741485.2A priority patent/EP2670863B1/en
Priority to ES12741485.2T priority patent/ES2687761T3/es
Publication of WO2012106385A2 publication Critical patent/WO2012106385A2/en
Publication of WO2012106385A3 publication Critical patent/WO2012106385A3/en
Priority to US15/597,917 priority patent/US10626442B2/en
Priority to US16/147,250 priority patent/US10982256B2/en
Priority to US16/163,486 priority patent/US10982271B2/en
Priority to US16/250,974 priority patent/US10995362B2/en
Priority to US16/691,493 priority patent/US20200157603A1/en
Priority to US16/795,203 priority patent/US20200208197A1/en
Priority to US16/808,100 priority patent/US11560585B2/en
Priority to US17/071,917 priority patent/US20210040538A1/en
Priority to US17/204,325 priority patent/US20220056515A1/en
Priority to US17/870,697 priority patent/US11566278B2/en
Priority to US17/870,641 priority patent/US11512341B1/en
Priority to US17/951,003 priority patent/US11634752B2/en
Priority to US17/951,013 priority patent/US11667956B2/en
Priority to US17/951,008 priority patent/US11732290B2/en
Priority to US18/086,369 priority patent/US11708599B2/en
Priority to US18/086,383 priority patent/US11692214B2/en
Priority to US18/102,056 priority patent/US11859240B2/en
Priority to US18/134,003 priority patent/US11781171B1/en
Priority to US18/233,214 priority patent/US11932902B2/en
Priority to US18/233,221 priority patent/US11932903B2/en
Priority to US18/379,589 priority patent/US11939624B2/en
Priority to US18/379,586 priority patent/US11926864B1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6806Preparing nucleic acids for analysis, e.g. for polymerase chain reaction [PCR] assay
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6816Hybridisation assays characterised by the detection means
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/686Polymerase chain reaction [PCR]

Definitions

  • the invention relates generally to the field of detection, identification, and quantification of target molecules in a sample.
  • the present invention relates in part to the detection, identification, and quantification of individual target molecules in single cells of a complex cell population while retaining cell specific information regarding that target molecule.
  • the invention relates to methods for identifying whether a plurality of targets are in a plurality of cells comprising: binding to the targets a plurality of tags, wherein a tag comprises a code that represents a) the target identity and b) the identity of the cell in which tag is binding.
  • individual cell separation or isolation is unnecessary for the binding step.
  • tags comprise building blocks that are directly or indirectly associated with each other, for example through covalent binding or by association through affinity. In some embodiments, tags are formed through polymerization of building blocks in place. In some embodiments, multiple building blocks are added in a step. In some embodiment, a single building block is added at each step. In some embodiments, the cell is alive. In some embodiments, the cell is lysed or fixed.
  • the invention relates to methods for identifying a single cell associated with a target comprising: binding to the target a tag, wherein the tag comprises a code that represents a) the target, and b) the single cell; wherein the during the binding the single cell is not isolated from a population of cells, and wherein the code that represents the single cell is unknown before the binding.
  • the invention relates to methods for identifying a single cell associated with a target comprising: binding to the target a tag, wherein the tag comprises a code that represents a) the target, and b) the single cell; wherein the during the binding the single cell is isolated from a population of cells, and wherein the code that represents the single cell is unknown before the binding.
  • the invention further comprises detecting the code, wherein individual cell separation or isolation is unnecessary for the detecting step.
  • each target is a protein or a nucleic acid.
  • the tag is a nucleic acid or a polypeptide.
  • the tag is comprises a series monomeric subunits that comprise a decipherable code.
  • the tag is a coded molecular constituent that can be decoded.
  • the tag comprises a combination of parts that can be decoded to determine the nature of the tag.
  • the tag comprises a UBA.
  • the UBA is specific for one of the targets.
  • the tag comprises a UBA.
  • the UBA comprises an antibody.
  • the tag comprises a ESB.
  • the ESB comprises a common linker (CL).
  • the ESB codes the target identity.
  • the ESB comprises a nucleic acid.
  • the tag comprises an APS.
  • the APS is detectable a detectably distinct coding unit. In some embodiments, during the binding step multiple APSs are added to the tag in an ordered manner during successive rounds of split pool synthesis.
  • the tag comprises at least 10 APSs.
  • the APS comprises a nucleic acid.
  • the tag comprises multiple APSs, an ESB, and a UBA linked by ligation.
  • multiple APSs, the ESB, and/or the UBA is capable of being linked Click chemistry.
  • the APS or the ESB comprises an amplification primer binding region.
  • the UBA, ESB, or APS is templatable.
  • the UBA, ESB, or APS is of a different discemable constituent (GPN: meaning one part of the code can be a nucleic acid, another can be a polypeptide, another can be a small molecule, etc.).
  • the invention relates to compositions comprising: a) a first target molecule, b) a first unique binding agent (UBA) specific for the first target molecule, c) a first linkable UBA-dependent epitope specific barcode (ESB), and d) a plurality of ordered assayable polymer subunit (APS), wherein the order of APSs is detectable.
  • UBA unique binding agent
  • EMB linkable UBA-dependent epitope specific barcode
  • APS ordered assayable polymer subunit
  • the target molecule is selected from the group consisting of a peptide, a polypeptide, an oligopeptide, a protein, a phosphoprotein, an antibody, a nucleic acid, a peptide nucleic acid, a synthetic small molecule, a disaccharide, a trisaccharide, an oligosaccharide, a polysaccharide, a lipid, a steroid, and a phospholipid.
  • the invention relates to compositions comprising a population of particles each comprising at least a first target molecule, wherein the first target molecule is associated with: a) a first unique binding agent (UBA) specific for the first target molecule, b) a first linkable UBA-dependent epitope specific barcode (ESB), and c) a first plurality of ordered assayable polymer subunits (APS), wherein the plurality of ordered APSs associated with the first target molecule of a first particle in the population is detectably different than the plurality of ordered APSs associated with the first target molecule of a second particle in the population.
  • UBA unique binding agent
  • ESD linkable UBA-dependent epitope specific barcode
  • the plurality of ordered APSs comprises 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 APSs. In some embodiments, the plurality of ordered APSs comprises more than 20 APSs. In some embodiments, the APSs are templatable. In some embodiments, at least one discrete particle is selected from the group consisting of a cell, a liposome, an organelle, a micelle, a droplet and a bead.
  • the target molecule is selected from the group consisting of a peptide, a polypeptide, an oligopeptide, a protein, a phosphoprotein, an antibody, a nucleic acid, a peptide nucleic acid, a synthetic small molecule, a disaccharide, a trisaccharide, an oligosaccharide, a polysaccharide, a lipid, a steroid, and a phospholipid.
  • the first ESB comprises a first common linker (CL).
  • said first target molecule is directly bound to said first UBA and said first ESB is directly bound to said first UBA.
  • the plurality of ordered APS is formed by stepwise addition of APSs in separate rounds.
  • the APS added on each round is linked to the first complex.
  • the linking is in order of rounds.
  • the linking is performed through binding affinity.
  • the linking of an APS, an ESB, or a UBA is performed using chemical methods.
  • the chemical method comprises Click chemistry.
  • the linking is performed in the presence of Cu 1 .
  • a UBA, an APS, or an ESB comprises nucleic acids.
  • oligonucleotide comprising a first and a second complementary region to two components selected from a UBA, an APS, and an ESB.
  • a UBA, an APS, or an ESB is linked using a linking oligonucleotide comprising the first and the second complementary region to two components selected from a UBA, an APS, and an ESB.
  • Some embodiments further comprise a second linking oligonucleotide comprising a third and a fourth complementary region to two components selected from a UBA, an APS, and an ESB.
  • a UBA, an APS, or an ESB is linked using a linking oligonucleotide comprising the third and the fourth complementary region to two components selected from a UBA, an APS, and an ESB.
  • the second and fourth complementary regions are identical.
  • the second and fourth complementary regions are identical.
  • the first or second complementary region is shared between two APSs within the plurality of APSs.
  • the linking is performed by ligation. In some embodiments, the linking
  • oligonucleotide comprises a subcode encoding the origin of the APS or the ESB.
  • the APS has a subcode encoding the origin of the APS.
  • the ESB has a subcode encoding the origin of the ESB.
  • an individual APS, ESB, or linking oligonucleotide molecule comprises a unique counter tag. In some embodiments, unique counter tag is detectable.
  • the ESB is covalently linked to the linking oligonucleotide.
  • an APS or an ESB comprises an amplification primer binding region.
  • the APSs and ESB when linked, are capable of encoding a secondary product.
  • the secondary product is an R A or a peptide.
  • the APSs and ESB when linked, comprises a polymerase start site.
  • the peptide comprises an affinity tag.
  • the affinity tag is a His-tag.
  • the UBA, ESB, or APS is templatable.
  • the composition further comprises a probe.
  • the probe is attached to a surface.
  • the surface comprises an array.
  • the surface comprises a bead.
  • the UBA is selected from the group consisting of antibody, peptide, aptamer, peptoid and nucleic acid.
  • the ESB is selected from the group consisting of nucleic acids, beads and chemical subunits.
  • said APS comprises a nucleic acid, a small molecule, or buildable complex molecules of deterministic weight.
  • kits for labeling a target molecule of a cell in a population of cells with a cell origination barcode comprising a) n sets of m assayable polymer subunits (APSs) each comprising a distinct package of information; wherein the packages of information are capable of being linked in an ordered fashion; b) a target molecule-specific unique binding agent (UBA).
  • APSs assayable polymer subunits
  • UAA target molecule-specific unique binding agent
  • kits for labeling a target molecule of a cell in a population of cells with a cell origination barcode comprising a) n sets of m assayable polymer subunits (APSs) each comprising a distinct package of information; wherein the packages of information are capable of being linked in an ordered fashion; b) a plurality of target molecule-specific unique binding agents (UBA) each linked with a UBA-specific epitope specific barcode (ESB).
  • UAA target molecule-specific unique binding agents
  • kits for labeling a target molecule of a cell in a population of cells with a cell origination barcode comprising a) n sets of m assayable polymer subunits (APSs) each comprising a distinct package of information; wherein the packages of information are capable of being linked in an ordered fashion; b) a plurality of target molecule-specific unique binding agents (UBA); c) a plurality of UBA-specific epitope specific barcode (ESB), wherein each ESB is capable of linking with a designated UBA.
  • n is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • m is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20.
  • n is greater than 10.
  • m is greater than 20.
  • the first ESB comprises a first common linker (CL).
  • said ESB is capable of directly binding to said UBA.
  • said UBA is capable of directly binding to the target molecule.
  • at least two of the assayable polymer subunit (APS) sets are identical.
  • the APSs in a first set is linkable to the APSs in a second set.
  • the APSs in a first set is further linkable to the APSs in a second set in an ordered fashion.
  • an APS, an ESB, or a UBA is capable of being linked using chemical methods.
  • the chemical method comprises Click chemistry.
  • the presence of Cu 1 is required for linkage.
  • the kit components can assemble through affinity binding.
  • a UBA, an APS, or an ESB comprises nucleic acids.
  • the kits further comprise a first linking oligonucleotide comprising a first and a second complementary region to two components selected from a UBA, an APS, and an ESB.
  • the kits further comprise a second linking oligonucleotide comprising a third and a fourth
  • the complementary region to two components selected from a UBA, an APS, and an ESB.
  • the first and third complementary regions are identical.
  • the second and fourth complementary regions are identical.
  • an APS, an ESB, or a UBA is capable of being linked by ligation. In some embodiments, the linking
  • oligonucleotide comprises a subcode encoding the original set of the APS or the ESB.
  • the APS has a subcode encoding the origin population of the APS.
  • the ESB has a subcode encoding the origin population of the ESB.
  • an individual APS, ESB, or linking oligonucleotide molecule comprises a unique counter tag.
  • the unique counter tag is detectable.
  • the ESB is covalently linked to the linking oligonucleotide.
  • an APS or an ESB comprises an amplification primer binding region.
  • the APSs and ESB when linked, are capable of encoding a secondary product.
  • the secondary product is an RNA or a peptide.
  • the APSs and ESB when linked, comprises a polymerase start site.
  • the peptide comprises an affinity tag.
  • the affinity tag is a His-tag.
  • the UBA, ESB, or APS is templatable.
  • the kit further comprises a probe.
  • the probe is attached to a surface.
  • the surface comprises an array.
  • the surface comprises a bead.
  • the plurality of UBAs comprises 2, 3, 4, 5, 10, 20, 30, 50, 100, 200, 300, 500, 600, 700, 800, 900, 1000 or more than 1000 UBAs. In some embodiments, the plurality of UBAs comprises up to 2000 UBAs. In some embodiments, the UBA is selected from the group consisting of antibody, peptide, aptamer, peptoid and nucleic acid. In some embodiments, the ESB is selected from the group consisting of nucleic acids, beads and chemical subunits. In some embodiments, said APS comprises a nucleic acid, a small molecule, or buildable complex molecules of deterministic weight.
  • the invention relates to methods for identifying target molecules sharing a common particle origin, comprising labeling a first plurality of targets of a first particle in a population of x particles with a first origination barcode; and labeling a second plurality of targets of a second particle in a population of x particles with a second origination barcode;
  • each origination barcode comprises a set of n assayable polymer subunits (APS);
  • x is greater than 1,000,000.
  • c is greater than 99.9%.
  • c is greater than 99.99%.
  • c is greater than 99.999%.
  • c is greater than 99.9999%.
  • c is greater than 99.99999%).
  • n is 2, 3, 4, 5, 6, 7, 8, 9, or 10.
  • m is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20.
  • n is greater than 10.
  • m is greater than 20.
  • At least one discrete particle is selected from the group consisting of a cell, a liposome, an organelle, a micelle, a droplet and a bead.
  • the target molecule is selected from the group consisting of a peptide, a polypeptide, an oligopeptide, a protein, a phosphoprotein, an antibody, a nucleic acid, a peptide nucleic acid, a synthetic small molecule, a disaccharide, a trisaccharide, an oligosaccharide, a polysaccharide, a lipid, a steroid, and a phospholipid.
  • at least two groups comprising m different APSs are identical.
  • n APSs are added in separate rounds.
  • the APSs of separate rounds are linked.
  • the linking is in order of rounds.
  • an appropriate n and/or m is selected based in a desired certainty level given a number of cells, x.
  • the invention relates to methods of imparting a particle specific code to a component of a particle of a population of particles, the method comprising: linking a first ordered set of assayable polymer subunits (APS) to a first component of a first particle of a population of particles, wherein the order of the APSs is detectable.
  • the method further comprises detecting the first ordered set of APSs linked with the first component, thereby determining a particle origin of the first component.
  • the method further comprises linking a second ordered set of assayable polymer subunits (APS) to a second component of the first particle of a population of particles, wherein the order of the APSs is detectable.
  • the method further comprises detecting the second ordered set of APSs linked with the second component, thereby determining the particle origin of the second component.
  • the first and the second ordered sets of APSs linked with the first and second components of the first particle are the same.
  • the method further comprises linking a third ordered set of assayable polymer subunits (APS) to a first component of second particle of a population of particles, wherein the order of the APSs is detectable.
  • the first ordered set of APSs linked with the first component of the first particle is different than the third ordered set of assayable polymer subunits linked with the first component of the second particle.
  • the method further comprises linking a component specific epitope specific barcode (ESB) to the first component. In some embodiments, the method further comprises linking a component specific ESB to the second component.
  • at least the particle is selected from the group consisting of a cell, a liposome, an organelle, a micelle, a droplet and a bead.
  • said at least one target molecule is directly bound to said first UBA and said ESB is directly bound to said UBA.
  • at least two of the assayable polymer subunit (APS) sets are identical. In some embodiments, each APS from the ordered set of APSs is linked to the first complex. In some embodiments, the linking is in order of rounds.
  • the UBA, ESB, or APS encodes a secondary product.
  • the secondary product is an RNA or a peptide.
  • the UBA, ESB, or APS is templatable.
  • the ESB further comprises a unique counter tag.
  • the quantity of the target molecule of the molecule is estimated using the counter tag.
  • the APS further comprises a round-specific subcode.
  • the detection further comprises determining the presence of an APS from a designated round.
  • detection is digital.
  • detection is indirect.
  • detecting comprises mass spectrometry.
  • detecting comprises nucleic acid sequencing.
  • detecting comprises peptide sequencing. In some embodiments, detecting comprises mass gel electrophoresis. In some embodiments, detecting comprises HPLC or other chromatographic separation. In some embodiments, detecting comprises detecting one or more signals associated with one or more individual APSs. In some embodiments, the signals are ordered. In some embodiments, detecting comprises using one or more probes. In some embodiments, the probe is attached to a surface. In some embodiments, the surface comprises an array. In some embodiments, the surface comprises a bead. In some embodiments, detecting comprises a separation. In some embodiments, the separation is multi-dimensional.
  • the separation resolves the first linkable UBA-dependent epitope specific barcode (ESB) from a second linkable UBA-dependent epitope specific barcode (ESB).
  • ESA linkable UBA-dependent epitope specific barcode
  • EMB linkable UBA-dependent epitope specific barcode
  • 3 4, 5, 10, 20, 30, 50, 100, 200, 300, 500, 600, 700, 800, 900, 1000 or more than 1000 different target molecules are detected. In some embodiments, up to 2000 different target molecules are detected.
  • the UBA is selected from the group consisting of antibody, peptide, aptamer, peptoid and nucleic acid.
  • the ESB is selected from the group consisting of nucleic acids, beads and chemical subunits.
  • said APS comprises a nucleic acid, a small molecule, or buildable complex molecules of deterministic weight.
  • the APSs are linked to through ligation or extension via polymerization.
  • a cell origination barcode (COB) is generated with the APSs from the ordered set of APSs.
  • COB cell origination barcode
  • each COB in said plurality of complexes has a detectable signal or sequence that distinguishes it from other COBs in said population of cells.
  • an APS, an ESB, or a UBA is linked using chemical methods.
  • the chemical method comprises Click chemistry.
  • the linking is performed in the presence of Cu 1 .
  • a UBA, an APS, or an ESB comprises nucleic acids.
  • the linking of a UBA, an APS, or an ESB is performed using a linking oligonucleotide that comprises a first and a second complementary region to two components to be linked.
  • the first or second complementary region is shared between APSs within a population of APSs.
  • the first or second complementary region is distinct for two different round-specific sets of APSs.
  • the method further comprises ligation.
  • the target molecule is selected from the group consisting of a peptide, a polypeptide, an oligopeptide, a protein, a phosphoprotein, an antibody, a nucleic acid, a peptide nucleic acid, a synthetic small molecule, a disaccharide, a trisaccharide, an oligosaccharide, a polysaccharide, a lipid, a steroid, and a phospholipid.
  • the first ESB comprises a first common linker (CL). In some embodiments, the first ESB comprises a first common linker (CL).
  • an individual APS, ESB, or linking oligonucleotide molecule comprises a unique counter tag.
  • detection comprises detecting the unique counter tag.
  • the number of unique counter tags associated with a specific ESB is determined.
  • the number of detected unique counter tags relate to the initial quantity of the specific ESB.
  • the ESB is covalently linked to the linking oligonucleotide.
  • an APS or an ESB comprises an amplification primer binding region.
  • a COB encodes a peptide sequence.
  • the COB comprises a polymerase start site.
  • the peptide comprises an affinity tag.
  • the affinity tag is a His-tag.
  • the invention relates to methods for detecting plurality of properties originating from a plurality of discrete particles, the method comprising: a) providing: i) a population of particles comprising at least a first target molecule; ii) a first unique binding agent (UBA) specific for the first target molecule; iii) a first linkable UBA-dependent epitope specific barcode (ESB); iv) a plurality of round-specific assayable polymer subunit (APS) sets, each set containing a plurality of APSs that are detectably distinct from each other; b) forming at least a first complex comprising said at least first target molecule, said first UBA probe, and said first ESB; c) performing n rounds of split pool synthesis, each round comprising; i) splitting the population of particles into m reaction volumes; ii) contacting one or more reaction volumes with an APS from the APS set
  • the invention relates to methods for detecting a plurality of properties originating from a plurality of discrete particles, the method comprising: a) providing: i) a population of particles comprising at least a first target molecule; ii) a first unique binding agent (UBA) specific for the first target molecule; iii) a first linkable UBA-dependent epitope specific barcode (ESB); and iv) a plurality of round-specific assayable polymer subunit (APS) sets, each set containing a plurality of APSs that are detectably distinct from each other; b) forming at least a first complex comprising said at least first target molecule, said first UBA probe, and said first ESB; c) performing n rounds of split pool synthesis, each round comprising: i) splitting the population of particles into m reaction volumes; ii) contacting one or more reaction volumes with an APS from the APS set specific for the round; and iii) pool
  • the split pool method is replaced by separation of particles, for example in microwells, or in microfluidic devices.
  • separated cells are labeled with cell origination barcodes.
  • cell origination barcodes are built in place by stepwise addition of building blocks.
  • n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20. In some embodiments, n is more than 20. In some embodiments, m is different between at least two rounds. In some embodiments, m is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20. In some embodiments, m is more than 20. In some embodiments, at least one discrete particle is selected from the group consisting of a cell, a liposome, an organelle, a micelle, a droplet and a bead. In some embodiments, the target molecule is selected from the group consisting of a peptide, a polypeptide, an oligopeptide, a protein, a
  • the first ESB comprises a first common linker (CL).
  • said at least one target molecule is directly bound to said first UBA and said ESB is directly bound to said UBA.
  • at least two of the assayable polymer subunit (APS) sets are identical.
  • the APS added on each round is linked to the first complex.
  • the linking is in order of rounds.
  • the UBA, ESB, or APS encodes a secondary product.
  • the secondary product is an RNA or a peptide.
  • the UBA, ESB, or APS is templatable.
  • the ESB further comprises a unique counter tag.
  • the quantity of the target molecule of the molecule is estimated using the counter tag.
  • the APS further comprises a round-specific subcode.
  • the detection further comprises determining the presence of an APS from a designated round.
  • detection is digital.
  • detection is indirect.
  • detecting comprises mass spectrometry.
  • detecting comprises nucleic acid sequencing.
  • detecting comprises peptide sequencing. In some embodiments, detecting comprises detecting one or more signals associated with one or more individual APSs. In some embodiments, the signals are ordered. In some embodiments, detecting comprises using one or more probes. In some embodiments, the probe is attached to a surface. In some embodiments, the surface comprises an array. In some
  • the surface comprises a bead.
  • detecting comprises a separation.
  • the separation is multi-dimensional.
  • the separation resolves the first linkable UBA-dependent epitope specific barcode (ESB) from a second linkable UBA-dependent epitope specific barcode (ESB).
  • ESD linkable UBA-dependent epitope specific barcode
  • 3 4, 5, 10, 20, 30, 50, 100, 200, 300, 500, 600, 700, 800, 900, 1000 or more than 1000 different target molecules are detected.
  • up to 2000 different target molecules are detected.
  • the UBA is selected from the group consisting of antibody, peptide, aptamer, peptoid and nucleic acid.
  • the ESB is selected from the group consisting of nucleic acids, beads and chemical subunits.
  • said APS comprises a nucleic acid, a small molecule, or buildable complex molecules of deterministic weight.
  • the APSs are linked to through ligation or extension via polymerization.
  • a cell origination barcode (COB) is generated from APSs of round specific APS sets.
  • each COB in said plurality of complexes has a detectable signal or sequence that distinguishes it from other COBs in said population of cells.
  • an APS, an ESB, or a UBA is linked using chemical methods.
  • the chemical method comprises Click chemistry.
  • the linking is performed in the presence of Cu 1 .
  • a UBA, an APS, or an ESB comprises nucleic acids.
  • the linking of a UBA, an APS, or an ESB is performed using a linking oligonucleotide that comprises a first and a second complementary region to two components to be linked.
  • the first or second complementary region is shared between APSs within a population of APSs.
  • the first or second complementary region is distinct for two different round-specific sets of APSs.
  • Some embodiments further comprise ligation.
  • the linking oligonucleotide comprises a subcode encoding the origin population of the APS or the ESB.
  • the APS has a subcode encoding the round-specific set of the APS.
  • the ESB has a subcode encoding the origin presence of the ESB.
  • an individual APS, ESB, or linking oligonucleotide molecule comprises a unique counter tag.
  • detection comprises detecting the unique counter tag.
  • the number of unique counter tags associated with a specific ESB is determined.
  • the number of detected unique counter tags relate to the initial quantity of the specific ESB.
  • the ESB is covalently linked to the linking oligonucleotide.
  • an APS or an ESB comprises an amplification primer binding region.
  • a COB encodes a peptide sequence.
  • the COB comprises a polymerase start site.
  • the peptide comprises an affinity tag.
  • the affinity tag is a His-tag.
  • each of the reaction volumes created by the most recent splitting receives a different APS from the APS set.
  • the invention provides methods for detecting at least one target molecule in a sample comprising the steps:(a) providing: (i) a population of cells potentially comprising at least one target molecule, (ii) a first UBA specific for a first target molecule, (iii) a first epitope specific barcode ESB specific for a region of the first UBA, where the ESB comprises a first common linker moiety, and (iv) a population of COB, where the population of COB comprises a second common linker moiety, where the second linker moiety is complementary to the first common linker moiety is the first ESB; (b) forming at least a first complex comprising the at least one target molecule, the first UBA probe, and the first ESB, where the at least one target molecule is bound to the first UBA and the ESB is bound to the UBA (c) adding the population of COBs, where a second complex is formed with the least one target molecule, the first
  • the invention provides methods for detecting at least one target molecule in a sample comprising the steps: (a) providing: (i) a population of cells potentially comprising at least one target molecule, (ii) a first unique binding agent (UBA) specific for a first target molecule, (iii) a first epitope specific barcode (ESB) specific for a region of the first UBA, where the ESB comprises a first common linker moiety, and (iv) a population of assayable polymer subunits (APSs), where the APSs comprises a second common linker moiety and a third common linker moiety, where the second linker moiety is complementary to the first common linker moiety is the first ESB; (b) forming at least a first complex comprising the at least one target molecule, the first UBA probe, and the first ESB, where the at least one target molecule is bound to the first UBA and the ESB is bound to the UBA; (c) splitting the
  • the methods further comprise detecting of a plurality of target molecules by forming a plurality of complexes in step (b), each complex comprising (i) at least one target molecule (ii) a first UBA and (iii) a first epitope specific barcode (ESB) specific for a region of the first UBA, where the ESB comprises a first common linker moiety, where the at least one target molecule is bound to the first UBA and the ESB is bound to the UBA.
  • ESB epitope specific barcode
  • each COB in the plurality of complexes has a detectable signal that distinguishes it from other COB in the population of cells.
  • the complex is detected by sequencing or mass spectrometry.
  • the third complex is detected by a method comprising individually counting the presence of one or more molecules of the third complex where the presence of the one or more molecules of the third complex is indicative of the concentration of the target molecule in a cell.
  • the individually detecting further comprises detecting a digital signal.
  • 3 4, 5, 10, 20, 30, 50, 100, 200, 300, 500, 600, 700, 800, 900, 1000 or more than 1000 different target molecules are detected. In some embodiments, up to 2000 different target molecules are detected.
  • the UBA is selected from the group consisting of antibody, peptide, aptamer, peptoid and nucleic acid.
  • the ESB is selected from the group consisting of nucleic acids, beads and chemical subunits.
  • the APS is a nucleic acid, a small molecule, or buildable complex molecules of deterministic weight. In some embodiments, the APS comprises a single-stranded nucleic acid hybridized to a complementary polynucleotide sequence having attached thereto a detectable label.
  • the first APS is attached to the first ESB through ligation or extension via polymerization.
  • the second APS is attached to the first APS through ligation or extension via polymerization.
  • the common linker moiety is a nucleic acid.
  • the ESB is attached to the USB.
  • said first COB comprises a plurality of APS.
  • Some embodiments further comprise detecting of a plurality of target molecules by a method comprising: forming a plurality of complexes in step (b), each complex comprising (i) at least one target molecule (ii) a first UBA and (iii) a first epitope specific barcode (ESB) specific for a region of said first UBA, wherein said ESB comprises a first common linker moiety, wherein said at least one target molecule is associated with said first UBA and said ESB is associated with said UBA.
  • a method comprising: forming a plurality of complexes in step (b), each complex comprising (i) at least one target molecule (ii) a first UBA and (iii) a first epitope specific barcode (ESB) specific for a region of said first UBA, wherein said ESB comprises a first common linker moiety, wherein said at least one target molecule is associated with said first UBA and said ESB is associated with said UBA.
  • each COB in said plurality of complexes has a detectable signal or sequence that distinguishes it from other COBs in said population of cells.
  • the linking of an APS, an ESB, or a UBA is performed using chemical methods.
  • the chemical method comprises Click chemistry.
  • the linking is performed in the presence of Cu 1 .
  • linking of an ABS, an ESB, or a UBA is performed using binding affinity.
  • a UBA, an APS, or an ESB comprises nucleic acids.
  • the linking of a UBA, an APS, or an ESB is performed using a linking oligonucleotide that comprises a first and a second complementary region to two components to be linked.
  • the first or second complementary region is shared between APSs within a population of APSs.
  • the first or second complementary region is distinct for different populations of APSs.
  • Some embodiments further comprise ligation.
  • the linking oligonucleotide comprises a subcode encoding the origin population of the APS or the ESB.
  • the APS has a subcode encoding the origin population of the APS.
  • the ESB has a subcode encoding the origin population of the ESB.
  • oligonucleotide molecule comprises a unique tag.
  • detection comprises detecting the unique tag.
  • the number of unique tags associated with a specific ESB is determined.
  • the number of detected unique tags relate to the initial quantity of the specific ESB.
  • the ESB is covalently linked to the linking oligonucleotide.
  • an APS or an ESB comprises an
  • a COB encodes a peptide sequence.
  • the COB comprises a polymerase start site.
  • the peptide comprises an affinity tag.
  • the affinity tag is a His-tag.
  • the two or more samples comprise at least 5 samples. In some embodiments, the two or more samples comprise at least 10 samples. In some embodiments, the two or more samples comprise at least 20 samples. In some embodiments, each of the samples created by the most recent splitting receives a different APS.
  • the invention relates to methods for labeling an ESB linked target molecule of a cell in a population of cells with a cell origination barcode (COB), comprising: separating each cell into an individual reaction volume; and adding the COB to the ESB via chemical or affinity means.
  • the reaction volume is selected from the group consisting of a microbubble, a microdroplet, a well, a microwell, and an enclosure in a microfluidics device.
  • the invention relates to methods comprising disassociating a variety of types of components originating from a cell and placing the components on a particle, wherein the components are labeled on said particle.
  • the labeling comprises labeling according to cell origin.
  • the labeling comprises labeling according to component type.
  • the signals in detection steps are ordered.
  • detecting comprises using one or more probes.
  • the probe is attached to a surface.
  • the surface comprises an array.
  • the surface comprises a bead.
  • detecting comprises a separation.
  • the separation is multi-dimensional.
  • the invention provides methods for preparing at least one UBA, ESB and/or APS as described herein.
  • the invention provides a population of UBAs, ESBs and/or APSs as described herein. In some embodiments, the invention provides kits comprising a population of UBAs, ESBs and APSs as described herein and instructions for its use.
  • Figure 1 depicts quantum of information representing a distinct signature (barcode) of the cell origin for each epitope.
  • Figure 2 shows a graphical representation of one embodiment of the components of the epitope specific barcode and cells origin barcodes of the invention and their assembly.
  • Figure 3 shows UBA-ESB-CL reagents of one embodiment of the invention.
  • Figure 4 depicts labeling of cells in one embodiment of the invention with UBA-ESB- CL reagents.
  • Figure 5 depicts ESB-COB reagents of one embodiment of the invention.
  • Figure 6 depicts ESB-COB assembly according to one embodiment of the invention.
  • Figure 7 depicts ESB-COB assembly according to another embodiment of the invention.
  • Figure 8 depicts ESB-COB assembly according to another embodiment of the invention.
  • Figure 9 depicts ESB-COB assembly according to another embodiment of the invention.
  • Figure 10 depicts ESB-COB assembly according to another embodiment of the invention.
  • Figure 11 depicts a peptide based ESB-COB readout according to one embodiment of the invention.
  • nucleic acid refers to a nucleotide polymer, and unless otherwise limited, includes known analogs of natural nucleotides that can function in a similar manner (e.g., hybridize) to naturally occurring nucleotides.
  • polynucleotide refers to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof.
  • Polynucleotides may have any three dimensional structure, and may perform any function, known or unknown.
  • polynucleotides coding or non-coding regions of a gene or gene fragment, intergenic DNA, loci (locus) defined from linkage analysis, exons, introns, messenger RNA (mRNA), transfer RNA, ribosomal RNA, short interfering RNA (siRNA), short-hairpin RNA (shRNA), micro-RNA (miRNA), small nucleolar RNA, ribozymes, complementary DNA (cDNA), which is a DNA representation of mRNA, usually obtained by reverse transcription of messenger RNA (mRNA) or by amplification; DNA molecules produced synthetically or by amplification, genomic DNA, recombinant polynucleotides, branched polynucleotides, plasmids, vectors, isolated DNA of any sequence, isolated RNA of any sequence, nucleic acid probes, and primers.
  • mRNA messenger RNA
  • transfer RNA transfer RNA
  • ribosomal RNA short interfering RNA
  • shRNA short-hairpin
  • a polynucleotide may comprise modified nucleotides, such as methylated nucleotides and nucleotide analogs. If present, modifications to the nucleotide structure may be imparted before or after assembly of the polymer. The sequence of nucleotides may be interrupted by non nucleotide components. A polynucleotide may be further modified after polymerization, such as by conjugation with a labeling component.
  • nucleic acid encompasses double- or triple-stranded nucleic acids, as well as single-stranded molecules. In double- or triple-stranded nucleic acids, the nucleic acid strands need not be coextensive (i.e., a double-stranded nucleic acid need not be double-stranded along the entire length of both strands).
  • nucleic acid also encompasses any chemical modification thereof, such as by methylation and/or by capping. Nucleic acid modifications can include addition of chemical groups that incorporate additional charge, polarizability, hydrogen bonding, electrostatic interaction, and functionality to the individual nucleic acid bases or to the nucleic acid as a whole. Such modifications may include base modifications such as 2'-position sugar
  • nucleic acids can include
  • nucleic acid that is an N- or C-glycoside of a purine or pyrimidine base
  • polyamide e.g., peptide nucleic acids (PNAs)
  • PNAs peptide nucleic acids
  • polymorpholino commercially available from the Anti- Virals, Inc., Corvallis, Oreg., as Neugene
  • nucleic acid also encompasses linked nucleic acids (LNAs), which are described in U.S. Pat. Nos.
  • the nucleic acid(s) can be derived from a completely chemical synthesis process, such as a solid phase-mediated chemical synthesis, from a biological source, such as through isolation from any species that produces nucleic acid, or from processes that involve the manipulation of nucleic acids by molecular biology tools, such as DNA replication, PCR amplification, reverse transcription, or from a combination of those processes.
  • a completely chemical synthesis process such as a solid phase-mediated chemical synthesis
  • a biological source such as through isolation from any species that produces nucleic acid, or from processes that involve the manipulation of nucleic acids by molecular biology tools, such as DNA replication, PCR amplification, reverse transcription, or from a combination of those processes.
  • a nucleic acid "probe” is an oligonucleotide capable of binding to a target nucleic acid of complementary sequence through one or more types of chemical bonds, generally through complementary base pairing, usually through hydrogen bond formation, thus forming a duplex structure.
  • the probe binds or hybridizes to a "probe binding site.”
  • the probe can be labeled with a detectable label to permit facile detection of the probe, particularly once the probe has hybridized to its complementary target. Alternatively, however, the probe may be unlabeled, but may be detectable by specific binding with a ligand that is labeled, either directly or indirectly. Probes can vary significantly in size. Generally, probes are at least 7 to 15 nucleotides in length.
  • probes are at least 20, 30, or 40 nucleotides long. Still other probes are somewhat longer, being at least 50, 60, 70, 80, or 90 nucleotides long. Yet other probes are longer still, and are at least 100, 150, 200 or more nucleotides long. Probes can also be of any length that is within any range bounded by any of the above values (e.g., 15-20 nucleotides in length).
  • a primer or probe can be perfectly complementary to the target nucleic acid sequence or can be less than perfectly complementary.
  • the primer has at least 65% identity to the complement of the target nucleic acid sequence over a sequence of at least 7 nucleotides, more typically over a sequence in the range of 10-30 nucleotides, and often over a sequence of at least 14-25 nucleotides, and more often has at least 75% identity, at least 85% identity, at least 90% identity, or at least 95%, 96%, 97%. 98%, or 99% identity.
  • certain bases e.g., the 3' base of a primer
  • Primer and probes typically anneal to the target sequence under stringent hybridization conditions.
  • binding interactions present in a mixture relying on affinity define specificity for two or more components' binding specificity.
  • a high binding affinity for a first interaction in comparison to binding affinities of other available interactions that are available for one or more binding partners in the first interaction will lead to high specificity.
  • Binding partners with high specificity form designated binding partners.
  • the invention provides methods, compositions and kits for detection and quantification of individual target molecules in biomolecular samples. In some embodiments, the invention provides methods, compositions and kits for detection and quantification of individual target molecules in every cell of a complex cell population while retaining cell specific information regarding that target molecule. Thus in some embodiments, the invention provides methods, compositions and kits for detection and quantification of individual target molecules in a single cell basis in samples with complex cell populations. Thus in some embodiments, for each cell the amount of each target molecule associated with that cell is assayed. In particular the invention provides unique binding agents that are capable of binding individual target molecules. The invention also provides the use of epitope specific barcode to tag target molecules.
  • the invention also provides the use of cell origination barcodes to indicate the cell of origin.
  • cell origination barcodes to indicate the cell of origin.
  • epitope specific barcodes and cell origination barcodes the binding of unique binding agents to target molecules results in the identification of the target molecules.
  • Methods of making and using such unique binding agents and/or epitope specific barcodes and/or cell origination barcodes are also provided.
  • the methods and compositions described herein can be used in a wide variety of applications such as diagnostic, prognostic, quality control and screening applications. Some embodiments of the invention relate to methods, compositions and kits for individually tagging cells.
  • epitope and "target molecule” are used interchangeably herein to refer to the molecule of interest (parts of it or the whole molecule) being detected and/or quantified by the methods described herein.
  • Certain aspects of the invention relate to the detection of multiple target molecules.
  • the methods described herein provide potential benefits in the areas of detection of multiple target molecules, quantification, and sensitivity.
  • the invention provides methods and compositions for the study of multiple protein measurements and/or multiple nucleic acid measurements that are sensitive and reliable.
  • Multiplexing within one sample at a single cell level is a key advantage of this approach. Multiplexing within one sample saves significant labor, reduces sample quantity requirements proportional to the number of measurements, and improves accuracy by elimination of errors compounded by separate sample handling and measurement steps. Furthermore, obtaining measurement of multiple target molecules in single cells in a complex cell population provides a better understanding of the physiological processes within each individual cell. In some embodiments, the methods described herein allow for the pooling of different samples together during processing to be analyzed at once. This offers throughput advantages and can accelerate the analysis of different samples.
  • the invention provides unique binding agents (UBA) for the analysis of target molecules.
  • UBA unique binding agents
  • the invention provides an UBA population for use in a multiplexed assay. Each UBA in the population is specific for a target molecule. Thus, the UBA provides the specificity for the target molecule recognized in a cell. The binding of the target molecules to the UBAs is then detected using epitope specific barcodes (ESB) and cell origination barcodes (COB). Each ESB comprises a unique code that can be associated to a specific target molecule. Each COB comprises a unique code that can be associated to a specific cell of origin.
  • ESB epitope specific barcodes
  • COB cell origination barcodes
  • the ESB are attached, directly or indirectly, to the UBA.
  • the ESBs bind to the UBAs in a cell or sample, e.g., as part of the assay procedure.
  • a unique COB is associated to the UBAs in a specific cell such that each COB can be associated to the target molecules bound to the UBAs in that cell.
  • the specific ESB/COB combination is referred as a quantum of information representing each target molecule or epitope (See Figure 1).
  • the COB is composed of one or more assayable polymer subunit (APS).
  • APS assayable polymer subunit
  • Certain aspects of the present invention relate to the selection of a library or population of designed (e.g., synthetic sequences) APS.
  • the present invention provides a population of designed (e.g. synthetic) APS wherein said APS comprises an unique sequence and/or a detectable molecule, and wherein the combination of one or more different APS in each COB has a detectable signal or sequence that distinguishes it from other COBs in said population.
  • the invention provides APSs comprising unique sequences (e.g. synthetic) that hybridize to a unique complementary polynucleotide sequence having attached thereto a detectable label.
  • the APS are detected by sequencing. Accordingly, certain aspects of the present invention provide a population of unique COBs or ESB/COBs, each comprised of a unique APS-based combination, wherein each COBs or ESB/COBs in the population is distinct from the other COBs or ESB/COBs in the population.
  • APSs generally are capable of forming a construct comprising the COBs. Any chemical structure allowing for such formation can be used for the APSs.
  • UBAs are molecules or assemblies that are designed to bind with at least one target molecule, at least one target molecule surrogate, or both; and can, under appropriate conditions, form a molecular complex comprising the UBA and the target molecule.
  • target molecules include, but are not limited to, proteins, nucleic acids, lipids, carbohydrates, ions, small molecules, organic monomers, and drugs.
  • target molecules include, but are not limited to, proteins, nucleic acids, lipids, carbohydrates, ions, small molecules, organic monomers, and drugs.
  • most of the embodiments described herein are explained in the context of UBAs that bind to a target protein or a target m NA. However, these embodiments also can be applied to other target molecules.
  • protein protein
  • polypeptide polypeptide
  • amino acid sequence amino acid sequence
  • the terms are used interchangeably herein to refer to polymers of amino acids of any length.
  • the polymer may be linear or branched, it may comprise modified amino acids, and it may be interrupted by non amino acids or synthetic amino acids.
  • the terms also encompass an amino acid polymer that has been modified, for example, by disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation, such as conjugation with a labeling component.
  • amino acid refers to either natural and/or unnatural or synthetic amino acids, including but not limited to glycine and both the D or L optical isomers, and amino acid analogs and peptidomimetics.
  • UBAs comprise at least one reaction portion that allow them to bind to or interact with at least one target molecule, at least one part of at least one target molecule, at least one target molecule surrogate, at least part of a target molecule surrogate, or combinations thereof;
  • the UBAs comprise an identity portion or at least part of an identity portion, for example, an ESB, a COB, an ESB and/or a linker oligo.
  • the UBAs comprise a capture region.
  • the capture region is used for the isolation of the UBA and/or immobilization of the UBA into a surface.
  • the capture region can be an affinity tag, a bead, a slide, an array, a microdroplet, an enclosure in a microfluidic device or any other suitable capture region in the art.
  • the capture region is the ESB, for example the ESB can be a detectable bead such as a bead with a unique spectral signature (e.g. a bead that has been internally dyed with red and infrared fluorophores).
  • Capture regions can define reaction volumes in which manipulation of compositions of the invention can take place.
  • the UBA is an antibody.
  • antibody and antibodies are used in a broad sense, to include not only intact antibody molecules, for example but not limited to immunoglobulin A, immunoglobulin G and immunoglobulin M, but also any immunoreactive component(s) of an antibody molecule that immunospecifically bind to at least one epitope.
  • immunoreactive components include but are not limited to, FAb fragments, FAb' fragments, FAb'2 fragments, single chain antibody fragments (scFv), miniantibodies, diabodies, crosslinked antibody fragments, AffibodyTM, cyclotides, molecules, and the like.
  • Immunoreactive products derived using antibody engineering or protein engineering techniques are also expressly within the meaning of the term antibodies.
  • Detailed descriptions of antibody and/or protein engineering, including relevant protocols, can be found in, among other places, J. Maynard and G. Georgiou, Ann. Rev. Biomed. Eng. 2:339 76 (2000); Antibody Engineering, R. Kontermann and S. Dubel, eds., Springer Lab Manual, Springer Verlag (2001); U.S. Pat. No. 5,831,012; and S. Paul, Antibody Engineering Protocols, Humana Press (1995).
  • antibody can be obtained from a variety of sources, including but not limited to polyclonal antibody, monoclonal antibody, monospecific antibody, recombinantly expressed antibody, humanized antibody, plantibodies, and the like; and can be obtained from a variety of animal species, including rabbit, mouse, goat, rat, human, horse, bovine, guinea pig, chicken, sheep, donkey, human, and the like.
  • a wide variety of antibodies are commercially available and custom-made antibodies can be obtained from a number of contract labs.
  • the antibodies described herein are attached to a nucleic acid, e.g., linker oligo or a nucleic acid ESB.
  • a nucleic acid e.g., linker oligo or a nucleic acid ESB.
  • Methods to attach nucleic acids to antibodies are known in the art. Any suitable method to attach nucleic acids to antibodies is encompassed in the methods of the invention.
  • the antibodies described herein can be attached to a nucleic acid by the methods described in GuUberg et al, PNAS 101 (22): pages 228420-8424 (2004); and Boozer et al, Analytical Chemistry, 76(23): pages 6967-6972 (2004), both incorporated herein by reference.
  • the antibodies described herein can be attached to a nucleic acid by random amine attachment.
  • the antibodies described herein can be attached to a nucleic acid by random amine attachment using a 10 to 1 ratio of nucleic acid to antibody.
  • the antibodies described herein can be attached to a nucleic acid by the methods described in Kozlov et al, Biopolymers 5: 73 (5): pages 621-630 (2004) incorporated herein by reference.
  • the antibodies described herein can be attached to a nucleic acid by hydrazine chemistry.
  • the antibodies described herein can be attached to a nucleic acid using tadpoles as described in Nolan, Nature Methods 2, 11 - 12 (2005), incorporated herein by reference.
  • the antibodies described herein can be attached to a nucleic acid by any suitable methods known in the art to generate engineered antibodies including the ones described herein.
  • the UBA is an aptamer.
  • Aptamers include nucleic acid aptamers (i.e., single-stranded DNA molecules or single-stranded RNA molecules) and peptide aptamers. Aptamers bind target molecules in a highly specific, conformation-dependent manner, typically with very high affinity, although aptamers with lower binding affinity can be selected if desired. Aptamers have been shown to distinguish between targets based on very small structural differences such as the presence or absence of a methyl or hydroxyl group and certain aptamers can distinguish between D- and L-enantiomers.
  • Aptamers have been obtained that bind small molecular targets, including drugs, metal ions, and organic dyes, peptides, biotin, and proteins, including but not limited to streptavidin, VEGF, and viral proteins. Aptamers have been shown to retain functional activity after biotinylation, fluorescein labeling, and when attached to glass surfaces and microspheres.
  • Nucleic acid aptamers are identified by an in vitro selection process known as systematic evolution of ligands by exponential amplification (SELEX).
  • SELEX systematic evolution of ligands by exponential amplification
  • oligonucleotides for example 1014 to 1015 individual sequences, often as large as 60 -100 nucleotides long, are routinely screened by an iterative process of in vitro selection and amplification.
  • Most targets are affinity enriched within 8 - 15 cycles and the process has been automated allowing for faster aptamer isolation.
  • Peptide aptamers are typically identified by several different protein engineering techniques known in the art, including but not limited to, phage display, ribosome display, mRNA display, selectively infected phage technology (SIP), and the like.
  • SIP selectively infected phage technology
  • nucleic acid aptamers and peptide aptamers can be obtained following conventional procedures and without undue experimentation. Detailed descriptions of aptamers, including relevant protocols, can be found in, among other places, L. Gold, J. Biol. Chem., 270(23): 13581 84 (1995); S.
  • the aptamer will be ligated or hybridized to nucleic acid such as a linker oligo or a nucleic acid ESB.
  • the hybridization or ligation of aptamers can be done by any suitable method known in art.
  • ligation can be performed enzymatically by at least one DNA ligase or at least one RNA ligase, for example but not limited to, T4 DNA ligase, T4 R A ligase, Thermus thermophilus (Tth) ligase, Thermus aquaticus (Taq) DNA ligase, or Pyrococcus furiosus (Pfu) ligase.
  • Ligation can also be performed by chemical ligation can, using activating and reducing agents such as carbodiimide, cyanogen bromide (BrCN), imidazole, 1- methylimidazole/carbodiimide/cystamine, N-cyanoimidazole, dithiothreitol (DTT) and ultraviolet light.
  • activating and reducing agents such as carbodiimide, cyanogen bromide (BrCN), imidazole, 1- methylimidazole/carbodiimide/cystamine, N-cyanoimidazole, dithiothreitol (DTT) and ultraviolet light.
  • the UBA is a peptoid.
  • Peptoids are short sequences of N- substituted glycines synthetic peptides that bind proteins.
  • small size peptoids improve diffusion and kinetics of the methods described herein. Any suitable method known in the art to generate peptoids is encompassed in the methods described herein. See Simon et al, PNAS 15; 89(20): 9367-9371 (1992), incorporated herein by reference.
  • the UBA is a nucleic acid sequence, e.g. an antisense DNA for a target mRNA.
  • the nucleic acid sequence is preferably at least 15 nucleotides in length, and more preferably is at least 20 nucleotides in length.
  • the target-specific sequence is about 10 to 500, 20 to 400, 30 to 300, 40 to 200, or 50 to 100 nucleotides in length. In other embodiments, the target-specific sequence is about 30 to 70, 40 to 80, 50 to 90, or 60 to 100, 30 to 120, 40 to 140, or 50 to 150 nucleotides in length.
  • the invention provides an epitope specific barcode (ESB).
  • ESB epitope specific barcode
  • Each ESB comprises a unique code that can be associated to a specific target molecule.
  • ESBs are molecules or assemblies that are designed to bind with at least one UBA or part of an UBA; and can, under appropriate conditions, form a molecular complex comprising the ESB, the UBA and the target molecule.
  • ESBs can comprise at least one identity identification portion that allow them to bind to or interact with at least one UBA; typically in a sequence-specific, a confirmation-specific manner, or both; for example but not limited to UBA-antibody binding, aptamer-target binding, and the like.
  • the ESB are attached, directly or indirectly, to the UBA.
  • the ESBs bind to the UBAs in a cell or sample, e.g., as part of the assay procedure.
  • the ESB is a solid surface or a capture region, for example, the ESB can be a detectable bead such as a bead with a unique spectral signature (e.g. a bead that has been internally dyed with red and infrared fluorophores).
  • the UBA is directly or indirectly attached to the capture region.
  • the ESBs comprise common linker moiety, for example, a linker oligo.
  • the common linker oligo is complementary to a common linker oligo in the assayable polymer subunits (APSs) that form the cell origination barcode (COB).
  • the ESBs comprise a capture region.
  • the capture region is used for the isolation of the ESB and/or immobilization of the ESB into a surface.
  • the capture region can be an affinity tag, a bead, a slide or an array.
  • the capture region is a detectable bead such as a bead with a unique spectral signature (e.g. a bead that has been internally dyed with red and infrared fluorophores
  • the ESB is an antibody or fragment thereof, an aptamer, a nucleic acid, or peptoid, as described above.
  • the ESB is a nucleic acid.
  • a part of the nucleic acid is amplified with branch chain or rolling circle approaches as known in the art.
  • the ESB is a peptoid.
  • Peptoids are short sequences of N- substituted glycines synthetic peptides that bind proteins.
  • small size peptoids improve diffusion and kinetics of the methods described herein. Any suitable method known in the art to generate peptoids is encompassed in the methods described herein. See Simon et al, PNAS 15; 89(20): 9367-9371 (1992), incorporated herein by reference.
  • the ESB is a nucleic acid sequence, e.g. an antisense nucleic acid for a complementary target nucleic acid sequence.
  • the nucleic acid sequence is preferably at least 15 nucleotides in length, and more preferably is at least 20 nucleotides in length.
  • the target-specific sequence is about 10 to 500, 20 to 400, 30 to 300, 40 to 200, or 50 to 100 nucleotides in length. In other embodiments, the target-specific sequence is about 30 to 70, 40 to 80, 50 to 90, or 60 to 100, 30 to 120, 40 to 140, or 50 to 150 nucleotides in length.
  • the invention provides a cell origination barcode (COB).
  • COB cell origination barcode
  • Each COB provides a unique code that can be associated to a specific cell of origin.
  • the COB code upon binding of the COB to a common linker moiety (e.g. common linker oligo) associated with an ESB, the COB code identifies the cells of origin of the target molecule to which the UBA/ESB complex is bound.
  • the COBs of the invention comprise two main portions: (i) a sequence specific for a common linker moiety (e.g. common linker oligo) associated with a UBA/ESB probe; and (ii) an unique code that can be associated to a specific cell of origin.
  • COBs are modular structures.
  • the COB comprises a plurality of different assayable polymer subunits (APS).
  • the COBs comprise a plurality of APSs attached in linear combination.
  • a COB is a molecular entity containing certain basic elements: (i) a plurality of APSs comprising label attachment regions attached in linear combination to form a backbone, and (ii)
  • the COBs comprise a plurality of different APSs attached in linear combination, wherein the APSs comprise small molecules of deterministic weight. In some embodiments, the COB comprises 2, 3, 4, 5, 6, 7, 8, 9, 10 or more unique APSs attached in a linear combination. In some
  • the COB comprises 4 or more APSs attached in linear combination.
  • the plurality of APSs attached in linear combination can comprise uniquely designed nucleic acid sequences.
  • the plurality of APSs attached in linear combination in the COBs can comprise at least one template, for example but not limited to, at least one nucleic acid sequence, such as at least part of a linear or linearizable viral genome, such as the genomes of adenovirus, hepatitis virus, herpes virus, rotavirus, and the like, or
  • bacteriophages such as lambda, Ml 3, ⁇ -174, T-series bacteriophages, and the like, including derivatives thereof comprising cloning cassettes, polylinkers, and the like; plasmids, such as pBR322 and pUC series plasmids, etc., including derivatives thereof comprising cloning cassettes, polylinkers, and the like; synthetic templates; templates comprising artificial sequences; and the like.
  • the skilled artisan will understand that virtually any piece of nucleic acid can serve as a template for fabricating a COB provided that it is large enough to include at least two APSs, or it can be combined with at least one other nucleic acid sequence so that the combined sequence is large enough to include at least two APSs.
  • ESBs, APSs, or COBs of the invention relate to templatable building blocks.
  • UBAs of the invention relate to templatable molecules.
  • the COB also comprises one or more APSs containing a common linker moiety (e.g. common linker oligo).
  • the common linker moiety can be directly or indirectly attached to the APSs.
  • the common linker moiety can be covalently attached to a COB or the common linker moiety can be bound to the COB later in the assay.
  • the term common linker moiety includes tandemly-repeated sequences of about 10 to about 25
  • the common linker moiety can be attached at either the 5' region or the 3' region of a COB, and may be utilized for capture and immobilization of a COB for imaging or detection, such as by attaching to a solid substrate a sequence that is complementary to the common linker moiety.
  • each molecular entity may be composed of one molecule or more than one molecule attached to one another by covalent or non-covalent means.
  • each component of a dual COB has a target molecule-specific UBA/ESB that binds to a different site on the same target molecule.
  • one of the COBs may be unlabeled.
  • the unlabeled COB may comprise a capture region.
  • COB is constructed from individual
  • the APSs are combined linearly.
  • the COB constructed from the APSs maintains the order of the APS.
  • COBs comprise branched structures.
  • the branched COB structures comprise information about the order of APS addition.
  • the individual APS forming a COB can be decoded.
  • the order or order of addition of the APSs forming the COB can be decoded. Without being bound by theory, platforms allowing the APS addition order to be protected and decoded lend for a higher number of different COBs to be generated from equivalent number of APS building blocks.
  • the methods of various embodiments of the invention allow for a higher statistical significance for the determination of cell/particle identity.
  • the complementary polynucleotide sequences attached to an APS serve to attach detectable molecules, or label monomers, to the APS.
  • complementary polynucleotide sequences may be directly labeled, for example, by covalent incorporation of one or more detectable molecules into the complementary polynucleotide sequence.
  • the complementary polynucleotide sequences may be indirectly labeled, such as by incorporation of biotin or other molecule capable of a specific ligand interaction into the complementary polynucleotide sequence.
  • the ligand e.g., streptavidin in the case of biotin incorporation into the complementary polynucleotide sequence
  • the ligand e.g., streptavidin in the case of biotin incorporation into the complementary polynucleotide sequence
  • the detectable molecules attached to an APS are not directly incorporated into the complementary polynucleotide sequence, this sequence serves as a bridge between the detectable molecule and the APS, and may be referred to as a bridging molecule, e.g., a bridging nucleic acid.
  • a bridging molecule e.g., a bridging nucleic acid.
  • COB/ESB/UBA complexes of the present invention comprise nucleic acids, which may be affinity-purified or immobilized using a nucleic acid, such as an oligonucleotide, that is complementary to a constant region of the COB (e.g. common linker moiety, capture region or affinity tag).
  • a nucleic acid such as an oligonucleotide
  • the COBs comprise at least one common linker moiety, which may serve as an affinity tag for purification and/or for immobilization (for example to a solid surface).
  • the common linker moiety can comprises two or more tandemly- repeated regions of repeat nucleotides, such as a series of 15-base repeats.
  • the COB whether complexed to an ESB, ESB/UBA, a target molecule/UBA/ESB or otherwise, can be purified or immobilized by an affinity reagent coated with a 15-base oligonucleotide which is the reverse complement of the repeat unit.
  • COBs, COB-ESB complexes, or COB/ESB/UBA complexes can be purified in two or more affinity selection steps.
  • the COB in the embodiments in which the COB is attached to an ESB/UBA complex, the COB can comprise an affinity tag.
  • one COB when a dual COB is used, one COB can comprise a first affinity tag and the other COB can comprise a second (different) affinity tag.
  • the COBs are mixed with the target molecules, and complexes comprising the two probes of the dual COBs are separated from unbound materials by affinity purification against one or both individual affinity tags.
  • the mixture can be bound to an affinity reagent for the first affinity tag, so that only probes comprising the first affinity tag and the desired complexes are purified.
  • the bound materials are released from the first affinity reagent and optionally bound to an affinity reagent for the second affinity tag, allowing the separation of complexes from COBs comprising the first affinity tag. At this point only full complexes would be bound.
  • the complexes are finally released from the affinity reagent for the second affinity tag and then analyzed by the methods described herein.
  • the affinity reagent can be any solid surface coated with a binding partner for the affinity tag, such as a column, bead (e.g., latex or magnetic bead) or slide coated with the binding partner.
  • affinity tags known in the art may be used, e.g., to purify and/or immobilize COBs, COB-ESB complexes, or COB/ESB/UBA complexes.
  • a biotin anchor is attached to the COB, ESB and/or UBA, allowing immobilization of the COBs, COB-ESB complexes, or COB/ESB/UBA complexes on a streptavidin surface (e.g. coated slide or bead).
  • an affinity tag is attached to a UBA, e.g., to purify and/or immobilize the UBA.
  • An affinity tag can be used for attachment to beads or other matrixes for a variety of useful applications including but not limited to purification. Examples of affinity tags and methods of making and/or attaching them to the nucleic acids are described in US patent 7,473,767; US applications No. 10/542,458; 12/324,357; 11/645,270 and 12/541,131, incorporated herein by reference in their entirety.
  • at least two of ESB, UBAs, APSs, and COBs comprise different chemical compositions described herein or any other suitable composition known in the art.
  • an ESB can comprise a peptide and/or a nucleic acid
  • an APS comprises a peptide or a peptoid. Any combination of described chemistries are envisioned within the scope of the invention.
  • ESBs, APSs and/or COBs can be template, ordered, and or decoded. In some embodiments, the order of any of these subunits in a construct can be detected.
  • ESBs, APSs and/or COBs can provide templates for the addition of any of another one of ESBs, APSs and/or COBs, for example via nucleic acid or any other suitable chemical complementarity known in the art.
  • ESBs, APSs and/or COBs may encode secondary products, such as a nucleic acid encoding for another nucleic acid or a peptide.
  • the detection of the ESBs, APSs and/or COBs comprises decoding the same, for example generating an amplicon or expressing a peptide from the ESBs, APSs and/or COBs and sequencing or otherwise detecting the products.
  • the sequences, the weights or the signals provided by the label monomers associated with the various APS of the COB of a given cell allow for the unique identification of the COB.
  • a COB having a unique identity or unique sequence signature is associated with a UBA that recognizes a specific target molecule or a portion thereof. Detection of the COB sequence allows for the detection of the presence of the target molecule in the mixture (qualitative analysis).
  • fluorescent labels a COB having a unique identity or unique spectral signature is associated with a UBA that recognizes a specific target molecule or a portion thereof.
  • Detection of the COB signal such as the spectral code of a fluorescently labeled COB allows detection of the presence of the target molecule in the mixture (qualitative analysis).
  • a COB having a unique deterministic weight is associated with a UBA that recognizes a specific target molecule or a portion thereof.
  • Detection of the COB deterministic weight allows detection of the presence of the target molecule in the mixture (qualitative analysis). Counting and or
  • UBA/ESB/COB complexes are thus useful for the diagnosis or prognosis of different biological states (e.g., disease vs. healthy) by quantitative analysis of known biological markers.
  • COBs of the invention allows for the identification of new diagnostic and prognostic markers, including those whose fluctuations among the different biological states is too slight to detect a correlation with a particular biological state using traditional molecular methods.
  • the sensitivity of COB-based molecular detection permits detailed pharmacokinetic analysis of therapeutic and diagnostic agents in small biological samples.
  • COB syntheses can be performed by any suitable methods known in the art, including the one described herein.
  • the invention relates to a COB to be synthesized by stepwise addition of assayable polymer subunits (APSs) comprising oligonucleotides.
  • APSs assayable polymer subunits
  • the COB can be attached to the UBA via a common linker (CL).
  • CL common linker
  • the CL can also be part of an
  • an epitope specific barcode is also provided as an oligonucleotide.
  • the epitope specific barcode can be included in the oligonucleotide that comprises the common linker.
  • CL, ESB and APSs all comprise oligonucleotide sequences. Accordingly, an oligonucleotide CL may be ligated to an oligonucleotide ESB or APS. Substantially complementary or exact complementary annealing regions may be utilized for hybridization. An annealing region may be provided on both ends of an oligonucleotide ESB or APS.
  • the APSs are added in various steps of a split pool synthesis or any other suitable stepwise synthesis known in the art.
  • An annealing region specific to each step of a stepwise synthesis maybe incorporated into the oligonucleotides.
  • the step- specific annealing region of the next oligonucleotide will not hybridize efficiently to the step- specific annealing region of the previous oligonucleotide that is available.
  • the some embodiments of the invention provide methods to stall the synthesis of COBs missing one or more APSs.
  • UBAs are labeled with different CL oligonucleotides, each with a unique ESB sequence specific to the UBA and a common annealing region.
  • the APSs are assembled into COBs during the rounds of split pool synthesis. In these cases, in each round, the sample is split into n different containers. A different oligonucleotide APS can be added to each container, totaling m different APSs. In some embodiments n and m are the same. In other embodiments, n is greater than m or m is greater than n.
  • Each APS can be designed so that it will selectively hybridize an annealing region added during the round before ( Figure 6).
  • a pair of round-specific annealing regions is incorporated in each APS.
  • the annealing regions can be incorporated to each end of the APS.
  • the annealing regions incorporated to each end of an APS can be different. Accordingly, an annealing region of an added APS can be complementary to an available annealing region from a previous round facilitating assembly.
  • the APSs are stitched together and/or to a CL using an annealing primer ( Figure 7 and 8).
  • the annealing primer may comprise a first complementary region to the CL or an APS added during the previous round of a stepwise synthesis.
  • the annealing primer may also comprise a second complementary region to the APS being added during a current round.
  • the annealing primer can hybridize to two oligonucleotide subunits of subsequent rounds stitching them together.
  • the first complementary regions of annealing primers of each round are different from the first complementary regions of annealing primers of other rounds ( Figure 7).
  • the second complementary regions of annealing primers of each round are different from the second complementary regions of annealing primers of other rounds. In some embodiments, the first or second complementary regions of annealing primers of different rounds are shared between rounds ( Figure 8).
  • a CL oligonucleotide comprises pairs of loop annealing regions ( Figures 9 and 10).
  • APSs can be designed to hybridize to the CL in a loop geometry, hybridizing on each end to the CL along the loop annealing regions.
  • the loop annealing regions can be specific to the round.
  • the hybridization can populate the APSs along the CL.
  • the APSs can be linked together using any method described herein or other common methods known in the art.
  • the APSs can be designed such that they do not efficiently hybridize to the CL along the loop annealing regions specific to other rounds. Consequently, if an APS from a particular round is missing, the APSs may not be linked together successfully, depending on the linking process.
  • a COB may be synthesized with a missing APS, the location of which is flanked by a pair of loop annealing regions. The resulting COB can then be analyzed accordingly and can either be discarded or the retrieved information can be
  • Each APS in a given round can comprise a unique subcode sequence that is different from the rest of the APSs in that round ( Figures 6-11).
  • the subcode may comprise a unique nucleotide sequence.
  • a CL, or one or more APSs may further comprise a random tag region allowing for subsequent normalization of the detected COBs ( Figures 6-11). Variations of suitable methods making use of such random tag regions are known in the art, e.g., see Casbon et al. (Nucleic Acids Research, 2011, 39: 12, e81).
  • the random tag region can function as a molecular counter to estimate the number of template molecules associated with each sequence variant.
  • a molecular counter is incorporated into a CL, ESB, APS or an assembled COB prior to an amplification reaction, e.g. PCR.
  • a library of molecular counters comprising degenerate base regions may be incorporated into CLs, ESBs, APSs or assembled COBs.
  • the number of unique DBRs in a library generally is limited by the length and base composition of the DBR. For example, a single nucleotide in a DBR would allow for four different possible counters, one for each base. Longer DBRs can produce higher numbers of unique counter sequences.
  • a molecular counter from a library of sequences can each be incorporated in a CL, ESB, APS or an assembled COB. The molecular counter can be used to determine whether a sequence variant is associated with a single template molecule, or alternatively, multiple template molecules.
  • the number of different DBR sequences associated with one sequence variant can serve as a direct measure of the number of initial template molecules. This information can supplement or replace the information provided by read numbers of each sequence variant, including, for example, read numbers after an amplification reaction, e.g. PCR. DBRs can also be used to determine the probability that a sequence variant derives from a polymerase error during an amplification reaction or is a true original variant prior to an amplification reaction, e.g. PCR. In some embodiments, unique binding agents (UBAs) are fixed to their targets prior to or concurrent with the assembly of COBs.
  • UBAs unique binding agents
  • COBs can, for example, be assembled associated with UBAs targeting cell surface components.
  • UBAs are fixed to cell surface components prior to or concurrent with the assembly of COBs.
  • UBAs are delivered into cells or into cellular compartments.
  • COBs are assembled associated with UBAs within cells or cellular compartments. Cells may be fixed prior to the addition of UBAs, ESBs or prior to COB assembly. Suitable cell permeabilization methods are known in the art and can be used to deliver components of the assay into cells and cellular components.
  • the assay is performed on bodies that are not cells.
  • Suitable support materials known in the art can act in the same manner a cell would act to provide an original binding surface.
  • Support materials can be decorated with binding targets.
  • support materials spatially resolve binding targets from each other.
  • the assay may comprise primary binding targets and one or more secondary binding targets that are capable of binding to the primary target.
  • a support material can for example, be coated with one or more primary targets.
  • a library of secondary targets can be provided to bind the primary targets.
  • UBAs can be provided to bind epitopes of primary and/or secondary targets.
  • COBs can be assembled associated with these UBAs as described for other types of targets. Inter-dependent binding of secondary targets to primary targets can be monitored by analyzing the COBs.
  • multiple COBs are assembled on the same UBA molecule.
  • the ESBs or assembled COBs encode a derivative sequence.
  • the ESBs and/or COBs comprise a polynucleotide sequence.
  • the ESB and/or COB can encode an R A sequence.
  • the ESB and/or the COB encode a peptide sequence.
  • the ESB and/or the COB can encode for the peptide sequence directly.
  • the COB can encode for a peptide sequence indirectly, for example, through an intermediary RNA sequence.
  • a polynucleotide ESB or COB can encode an open reading frame.
  • the peptide sequence is translated after introducing the ESB or COB into a construct enabling peptide expression.
  • the construct is a vector.
  • the ESBs and COBs are assembled from oligonucleotides.
  • the linking agent can be a ligase.
  • the ligase is T4 DNA ligase, using well known procedures (Maniatis, T. in Molecular Cloning, Cold Spring Harbor Laboratory (1982)).
  • Other DNA ligases may also be used.
  • ligation other ligases, such as those derived from thermophilic organisms may be used thus permitting ligation at higher
  • the linking agent may be a chemical agent which will cause the ESB and APSs to link unless there is a nucleotide base pair mismatching at the aneealing region.
  • T4 DNA ligase As the linking agent. This enzyme requires the presence of a phosphate group on the 5' end that is to be joined to a 3' OH group on a neighboring oligonucleotide.
  • oligos When oligos are stacking together to bind to an annealing region with a perfect match at the junction at their ends, it results in a specific binding to the annealing region.
  • the CLs, ESBs and APSs can be ligated to form an ESB linked COB.
  • the ESB linked COBs in turn, can be used for detection.
  • the ESB and/or COB assembly comprises the use of
  • CLICK chemistry Suitable methods to link various molecules using CLICK chemistry are known in the art (for CLICK chemistry linkage of oligonucleotides, see, e.g. El-Sagheer et al. (PNAS, 108:28, 11338-11343, 2011).
  • the ESB and/or COB assembly takes place inside a cell.
  • the ESBs and/or APSs are first assembled inside a cell.
  • the ESBs and/or APSs are linked inside a cell. In some embodiments, the ESBs and/or APSs are linked outside a cell.
  • the assembled products are amplified and, optionally, the results are compared with amplification of similar target nucleic acids from a reference sample.
  • the ligated products of APSs are amplified and, optionally, results are compared with amplification of a similar COB from a reference sample. Amplification can be performed by any means known in the art. In some cases, the ligated products are amplified by polymerase chain reaction (PCR).
  • PCR techniques include, but are not limited to, quantitative PCR, quantitative fluorescent PCR (QF-PCR), multiplex fluorescent PCR (MF-PCR), real time PCR (RTPCR), single cell PCR, restriction fragment length polymorphism PCR (PCR-RFLP), PCK-RFLPIRT-PCR-IRFLP, hot start PCR, nested PCR, in situ polonony PCR, in situ rolling circle amplification (RCA), bridge PCR, picotiter PCR and emulsion PCR.
  • QF-PCR quantitative fluorescent PCR
  • MF-PCR multiplex fluorescent PCR
  • RTPCR real time PCR
  • PCR-RFLP restriction fragment length polymorphism PCR
  • PCK-RFLPIRT-PCR-IRFLP PCK-RFLPIRT-PCR-IRFLP
  • LCR ligase chain reaction
  • transcription amplification self-sustained sequence replication
  • selective amplification of target polynucleotide sequences consensus sequence primed polymerase chain reaction (CP-PCR), arbitrarily primed polymerase chain reaction (AP-PCR), degenerate oligonucleotide-primed PCR (DOP-PCR) and nucleic acid based sequence amplification (NABSA).
  • CP-PCR consensus sequence primed polymerase chain reaction
  • AP-PCR arbitrarily primed polymerase chain reaction
  • DOP-PCR degenerate oligonucleotide-primed PCR
  • NABSA nucleic acid based sequence amplification
  • Other amplification methods that can be used herein include those described in U.S. Patent Nos. 5,242,794;
  • the amplification is performed inside a cell.
  • amplification of ligated products may occur on a support, such as a bead or a surface.
  • COBs may be assembled on targets from a single cell.
  • each UBA/ESB probe in a population of probes allows for the multiplexed analysis of a plurality of target molecules. Furthermore the uniqueness of each COB probe in a population of probes allows for the multiplexed analysis of a plurality of target molecules in single cells.
  • each COB contains six APSs. If the APSs are going to be sequenced and there are 20 possible unique sequences for the APS. There will be
  • 3.84 x 10 possible COBs in this example 3.84 x 10 cells and their corresponding UBA/ESB probes can be analyzed in this example.
  • the methods of detection are performed in multiplex assays, whereby a plurality of target molecules is detected in the same assay (a single reaction mixture).
  • the assay is a hybridization assay or an affinity binding assay in which the plurality of target molecules is detected simultaneously.
  • the assay is hybridization assay or an affinity binding assay in which the plurality of target molecules is detected simultaneously in single cells.
  • the plurality of target molecules detected in the same assay is, at least 2, at least 5 different target molecules, at least 10 different target molecules, at least 20 different target molecules, at least 50 different target molecules, at least 75 different target molecules, at least 100 different target molecules, at least 200 different target molecules, at least 500 different target molecules, or at least 750 different target molecules, or at least 1000 different target molecules.
  • the plurality of target molecules detected in the same assay is up to 50 different target molecules, up to 100 different target molecules, up to 150 different target molecules, up to 200 different target molecules, up to 300 different target molecules, up to 500 different target molecules, up to 750 different target molecules, up to 1000 different target molecules, up to 2000 target molecules, or up to 5000 target molecules.
  • the plurality of target molecules detected is any range in between the foregoing numbers of different target molecules, such as, but not limited to, from 20 to 50 different target molecules, from 50 to 200 different target molecules, from 100 to 1000 different target molecules, from 500 to 5000 different target molecules, and so on and so forth.
  • the detection is digital detection.
  • the detection is direct, i.e. the method acquires a signal that is directly generated by the detected entity.
  • the detection is indirect, i.e. manipulation of an entity to be detected takes place before a signal is acquired.
  • a plurality of components of an entity to be detected give rise to detection signals directly or indirectly.
  • the order of the plurality of the components can be determined through the detection methods described herein. Such detection methods are also described as ordered detection methods or detection methods with ordered signals.
  • the detection or quantification analysis of the COBs can be accomplished by sequencing.
  • the APS subunits or entire COBs can be detected via full sequencing of all DNA tags by any suitable methods known in the art, e.g., Illumina HiSeq 2000, including the sequencing methods described herein.
  • Sequencing can be accomplished through classic Sanger sequencing methods which are well known in the art. Sequencing can also be accomplished using high-throughput systems some of which allow detection of a sequenced nucleotide immediately after or upon its incorporation into a growing strand, i.e., detection of sequence in red time or substantially real time. In some cases, high throughput sequencing generates at least 1,000, at least 5,000, at least 10,000, at least 20,000, at least 30,000, at least 40,000, at least 50,000, at least 100,000 or at least 500,000 sequence reads per hour; with each read being at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 120 or at least 150 bases per read.
  • high-throughput sequencing involves the use of technology available by Illumina' s HiSeq 2000 machine. This machine uses reversible terminator-based sequencing by synthesis chemistry. This machine can do 200 billion DNA reads in eight days.
  • high-throughput sequencing involves the use of technology available by ABI Solid System. This genetic analysis platform that enables massively parallel sequencing of clonally-amplified DNA fragments linked to beads.
  • the sequencing methodology is based on sequential ligation with dye-labeled oligonucleotides.
  • high-throughput sequencing involves the use of technology available by Ion Torrent Personal Genome Machine (PMG).
  • PMG Personal Genome Machine
  • the PGM can do 10 million reads in two hours.
  • high-throughput sequencing involves the use of technology available by Helicos Biosciences Corporation (Cambridge, Massachusetts) such as the Single Molecule Sequencing by Synthesis (SMSS) method.
  • SMSS is unique because it allows for sequencing the entire human genome in up to 24 hours. This fast sequencing method also allows for detection of a SNP nucleotide in a sequence in substantially real time or real time.
  • SMSS is powerful because, like the MIP technology, it does not require a pre amplification step prior to hybridization. In fact, SMSS does not require any amplification. SMSS is described in part in US Publication Application Nos. 2006002471 I; 20060024678; 20060012793;
  • high-throughput sequencing involves the use of technology available by 454 Lifesciences, Inc. (Branford, Connecticut) such as the Pico Titer Plate device which includes a fiber optic plate that transmits chemiluninescent signal generated by the sequencing reaction to be recorded by a CCD camera in the instrument.
  • This use of fiber optics allows for the detection of a minimum of 20 million base pairs in 4.5 hours.
  • high-throughput sequencing is performed using Clonal Single Molecule Array (Solexa, Inc.) or sequencing-by-synthesis (SBS) utilizing reversible terminator chemistry.
  • Solexa, Inc. Clonal Single Molecule Array
  • SBS sequencing-by-synthesis
  • RNA or DNA can take place using AnyDot.chjps (Genovoxx, Germany).
  • AnyDot-chips allow for lOx - 50x enhancement of nucleotide fluorescence signal detection. AnyDot.chips and methods for using them are described in part in International Publication Application Nos. WO02/088382,
  • WO03/020968 WO03/031947, WO2005/044836, PCT/EP05/105657, PCT/EP05/105655; and German Patent Application Nos. DE 101 49 786, DE 102 14 395, DE 103 56 837, DE 10 2004 009 704, DE 10 2004 025 696, DE 10 2004 025 746, DE 10 2004 025 694, DE 10 2004 025 695, DE 10 2004 025 744, DE 10 2004 025 745, and DE 10 2005 012 301.
  • Sequence can then be deduced by identifying which base is being incorporated into the growing complementary strand of the target nucleic acid by the catalytic activity of the nucleic acid polymerizing enzyme at each step in the sequence of base additions.
  • a polymerase on the target nucleic acid molecule complex is provided in a position suitable to move along the target nucleic acid molecule and extend the oligonucleotide primer at an active site.
  • a plurality of labeled types of nucleotide analogs are provided proximate to the active site, with each distinguishably type of nucleotide analog being complementary to a different nucleotide in the target nucleic acid sequence.
  • the growing nucleic acid strand is extended by using the polymerase to add a nucleotide analog to the nucleic acid strand at the active site, where the nucleotide analog being added is complementary to the nucleotide of the target nucleic acid at the active site.
  • the nucleotide analog added to the oligonucleotide primer as a result of the polymerizing step is identified.
  • the steps of providing labeled nucleotide analogs, polymerizing the growing nucleic acid strand, and identifying the added nucleotide analog are repeated so that the nucleic acid strand is further extended and the sequence of the target nucleic acid is determined.
  • oligonucleotide ESB or COBs are identified directly.
  • the direct identification can comprise nucleic acid sequencing described supra.
  • the APS sequences encode for derivative sequences that are in turn identified.
  • a polynucleotide ESB and COB can be translated into a peptide sequence.
  • the peptide sequence can then be identified using suitable methods known in the art.
  • a sequence representing a COB or ESB is identified using mass spectrometric analysis. Sequencing methods comprising mass spectrometry are known in the art. In various embodiments, a derivative sequence, such as a peptide, is sequenced using mass spectrometric methods. In some embodiments, the mass spectrometric methods comprise fragmentation. In some embodiments, the mass spectrometric methods comprise N-terminal sequencing. In some embodiments, the mass spectrometric methods comprise C-terminal sequencing. In some embodiments, the mass spectrometric methods comprise Edman
  • the derivative sequence is subjected to a separation process prior to identification.
  • the separation process comprises chromatography.
  • the separation process comprises HPLC. Suitable separation methods for the separation process comprise, for example, ion-exchange
  • Ion-exchange chromatography can use any matrix material comprising functional groups that are strongly acidic (typically, sulfonic acid groups, e.g. sodium polystyrene sulfonate or polyAMPS), strongly basic, (quaternary amino groups, for example, trimethylammonium groups, e.g. polyAPTAC), weakly acidic (mostly, carboxylic acid groups), or weakly basic (primary, secondary, and/or ternary amino groups, e.g. polyethylene amine).
  • the separation method can, for example, use sulfonated polystyrene as a matrix, adding the amino acids in acid solution and passing a buffer of steadily increasing pH through the column.
  • Amino acids are be eluted when the pH reaches their respective isoelectric points.
  • Hydrophobic interaction chromatography may be employed through the use of reversed phase chromatography.
  • Many commercially available C8 and C18 silica columns have demonstrated successful separation of peptides in solution through the use of an elution gradient.
  • peptide sequences representing ESBs, APSs and the resulting COBs are designed to improve the ease of detection.
  • peptide sequences can be designed to improve fragmentation patterns in a mass spectrometer. It is well known in the art that the fragmentation efficiency of a bond in a peptide sequence is sequence dependent (see, e.g. Tabb et al. Anal Chem. 2003 March 1; 75(5): 1155 and Klammer et al. Bioinformatics 2008, 24: 348-356). The sequence-dependent fragmentation efficiencies can be employed to design representative peptide sequences with desired fragmentation patterns.
  • peptide sequences representing ESBs, APSs and the resulting COBs are designed to confer certain physical and chemical characteristics to the peptide molecules.
  • representative peptide sequences can be designed to result in peptide molecules with solubilities in aqueous solutions within a desired range.
  • representative peptide sequences can be designed to result in peptide molecules with a desired degree of secondary or tertiary structure or lack thereof.
  • representative peptide sequences can be designed to result in peptide molecules with disulfide bonds or lack thereof.
  • representative peptide sequences can be designed to result in peptide molecules with desired binding characteristics to chosen targets.
  • sequences of the ESBs, CLs, APSs and the resulting COBs can further be designed to exploit specially loaded tRNAs in a protein expression system. Accordingly, incorporation of non-natural amino acids to the resulting peptide molecules can be accomplished.
  • Various embodiments relate to separating the ESB-linked COBs or derivative sequences, such as a peptide sequence, prior to detection.
  • the separation is based on a suitable physiochemical property of the molecules.
  • These types of separations are particularly useful to direct the molecules to the detectors sequentially thereby increasing their relative abundance and the complexity of the signal at the time of the detection.
  • Various embodiments comprise separation of the molecules in a sequence-targeted manner. For example, all molecules comprising a particular ESB can be isolated by hybridizing the ESB sequence to a sufficiently complementary sequence, affinity purifying using an ESB-specific linked tag, affinity purifying using a derivative sequence encoded by the ESB sequence or any other suitable method known in the art. Separation methods may also target a cell-specific COB or a portion thereof.
  • constructs comprising ESBs and COBs are subjected to separation.
  • the constructs can be subjected to gradient or affinity purification sorting according to the ESBs.
  • the separation may comprise multiple dimensions, for example 2, 3, 4, 5, 6, 7, or more dimensions.
  • the constructs can be separated sorting according to the first APSs on one dimensions and sorting the according to the second APSs on another dimension and optionally sorting according to the ESBs on a third dimension.
  • the separation method comprises separation by gradient, on a gel, using an electromagnetic field and/or any other suitable separation method known in the art.
  • constructs comprising ESBs and/or COBs can be separated prior to detection.
  • the constructs can be separated according to ESBs and the ESBs and/or COBs in the constructs can be detected following the separation.
  • the detection can be molar ratio detection, enzymatic detection, sequencing, differential affinity in a gradient or gel, electromagnetic field, e.g. UV, fluorescence or chemiluminescence, any detection method described herein, or any other suitable detection method known in the art.
  • the separation comprises immobilizing the constructs.
  • the constructs can be immobilized on an array surface or on a bead.
  • a portion of the ESB and/or COB can be used to immobilize the constructs.
  • the ESB and/or the COB can be detected from immobilization positions.
  • ESBs and/or COBs comprising oligonucleotide can be immobilized on a bead or microarray that is coated with complementary oligonucleotides.
  • the COBs of the present invention can be labeled with any of a variety of label monomers, such as a radioisotope, fluorochrome, dye, enzyme, nanoparticle, chemiluminescent marker, biotin, or other monomer known in the art that can be detected directly (e.g., by light emission) or indirectly (e.g., by binding of a fluorescently-labeled antibody).
  • label monomers such as a radioisotope, fluorochrome, dye, enzyme, nanoparticle, chemiluminescent marker, biotin, or other monomer known in the art that can be detected directly (e.g., by light emission) or indirectly (e.g., by binding of a fluorescently-labeled antibody).
  • labeled APSs in the COB is labeled with one or more label monomers, and the signals provided by the label monomers attached to the APS of a COB constitute a detectable code that identifies the target to which the UBA the COB binds.
  • Example of label monomers that can be used with the COBs described herein and methods to incorporate the labels monomers into the COBs are described in US patent 7,473,767; US applications No. 10/542,458; 12/324,357; 11/645,270 and 12/541,131, incorporated herein by reference in their entirety.
  • the COB of the invention are uniquely suitable for conducting quantitative analyses.
  • the COB of the invention By providing a one to one binding between the COB of the invention and their target molecules in a biomolecular sample, all or a representative portion of the target molecules present in the sample can be identified and counted.
  • This individual counting of the various molecular species provides an accurate and direct method for determining the absolute or relative concentration of the target molecule in the biomolecular sample.
  • the ability to address each molecule in a mixture individually leverages benefits of miniaturization including high sensitivity, minimal sample quantity requirements, high reaction rates which are afforded by solution phase kinetics in a small volume, and ultimately very low reagent costs.
  • Target molecules or epitopes are the molecules detected or measured by binding of a UBA whose target-specific region(s) recognize thereto.
  • target molecules include, but are not limited to, proteins, nucleic acids, lipids, carbohydrates, small molecules, organic monomers, or drugs.
  • Nucleic acids that can be analyzed by the methods herein include: double-stranded DNA, single-stranded DNA, single-stranded DNA hairpins, DNA/RNA hybrids, RNA (e.g. mRNA or miRNA) and RNA hairpins.
  • RNA e.g. mRNA or miRNA
  • the methods described herein are explained mostly in the context of analyzing proteins or mRNA. However, the embodiments described herein also can be used to detect non-protein or non-mRNA targets.
  • the target molecule is selected from the group consisting of a peptide, a polypeptide, an oligopeptide, a protein, a phosphoprotein, an antibody, a nucleic acid, a peptide nucleic acid, a synthetic small molecule, a disaccharide, a trisaccharide, an oligosaccharide, a polysaccharide, a lipid, a steroid, and a phospholipid.
  • a target molecule can be part of a biomolecular sample that contains other components or can be the sole or major component of the sample.
  • a target molecule can be a component of a whole cell or tissue, a cell or tissue extract, a fractionated lysate thereof or a substantially purified molecule.
  • the target molecule can be attached in solution or solid-phase, including, for example, to a solid surface such as a chip, microarray or bead. Also the target molecule can have either a known or unknown structure or sequence.
  • the compositions, methods, and kits disclosed herein can also be used in a wide variety of applications to determine the presence of target molecules in a sample.
  • compositions, methods, and kits are useful for, pharmacokinetic studies, including but not limited to, drug metabolism, ADME profiling, and toxicity studies; target validation for drug discovery; gene expression profiling, protein expression profiling; proteome analyses; metabolomic studies; post-translation modification studies, including but not limited to glycosylation, phosphorylation, acetylation, and amino acid modification, such as modification of glutamate to form gamma-carboxy glutamate and hydroxylation of proline to form hydroxylation; analyses of specific serum or mucosal antibody levels; evaluation of non- nucleic acid diagnostic indicators; foreign antigen detection; and the like.
  • At least one UBA, at least one ESB, or both the UBA and the ESB comprise at least one antibody, aptamer or peptoid that reacts specifically with at least one target molecule.
  • at least one UBA, at least ESB, or both the UBA and the ESB comprise binding proteins that specifically interact with at least one target molecule.
  • the ESB comprise a common linker moiety.
  • molecular complexes and the at least part of the molecular complexes described herein can be individually detected while tethered or attached to a substrate or while in solution, depending on, among other things, the nature of the specific molecular complex or cleavable component and the SMD technique and detection apparatus employed.
  • the present invention provides methods for detection and quantification of target molecules in biomolecular samples.
  • the invention provides UBAs that are capable of binding individual target molecules.
  • the invention also provides the use of ESBs and COBs (See Figure 2). Through the ESBs' and COBs' codes, the binding of the UBAs to target molecules results in the identification of the target molecules in single cells.
  • ESBs and COBs' codes Through the ESBs' and COBs' codes, the binding of the UBAs to target molecules results in the identification of the target molecules in single cells.
  • the ESB/COB complex represents a quantum of information that represents the target molecule and the cell of origin (See Figure 1). Methods of making and using such UBAs and/or ESBs and COBs are also provided.
  • the invention provides methods to identify multiple target molecules in every cell of a complex cell population and to retain cell specific information regarding that target molecule. Therefore, for each cell the amount of each target molecule associated with that cell is assayed. In some embodiments, multiple quanta of information are determined to identify multiple target molecules in every cell of a complex cell population.
  • the invention provides methods for detecting at least one target molecule in a sample comprising the steps:(a) providing: (i) a population of cells potentially comprising at least one target molecule, (ii) a first UBA specific for a first target molecule, (iii) a first epitope specific barcode ESB specific for a region of the first UBA, where the ESB comprises a first common linker moiety, and (iv) a population of COB, where the population of COB comprises a second common linker moiety, where the second linker moiety is complementary to the first common linker moiety is the first ESB; (b) forming at least a first complex comprising the at least one target molecule, the first UBA probe, and the first ESB, where the at least one target molecule is bound to the first UBA and the ESB is bound to the UBA (c) adding the population of COBs, where a second complex is formed with the least one target molecule, the first
  • the invention provides methods for detection and/or quantification of a target molecule by binding a UBA to a target molecule.
  • a UBA comprises at least one reaction portion that allow the UBA to bind to or interact with the target molecule; typically in a sequence-specific, a confirmation-specific manner, or both; for example but not limited to antigen-antibody binding, aptamer-target binding, and the like (See Figures 2 and 3).
  • UBAs can be part of at least one probe set, comprising at least one first probe and at least one second probe.
  • the invention provides methods for detection and/or quantification of a target molecule by binding a UBA probe set to a target molecule, where the UBA probe set comprises a first UBA probe and a second UBA probe.
  • the first UBA probe and the second UBA probe comprise at least one reaction portion that allow the probes to bind to or interact with different regions of the target molecule, e.g., in a sequence-specific manner, a confirmation-specific manner, or both.
  • the UBA probe and/or a second UBA probe contain a capture region as described herein.
  • the UBAs comprise an identity portion or at least part of an identity portion, for example, an ESB, a COB, an ESB and/or a linker oligo.
  • the identity portion allows for the identification of the presence or absence of the UBAs bound to the target molecule in the detection step of the methods described herein.
  • the invention provides methods for detection and/or quantification of a target molecule by binding the UBA to a target molecule, wherein UBA contains an identity portion (e.g., ESB, a COB, an ESB and/or a linker oligo).
  • the target molecule is tagged within cells indirectly with an epitope specific barcode (ESB).
  • Each ESB comprises a unique code that can be associated to a specific target molecule.
  • ESBs are molecules or assemblies that are designed to bind with at least one UBA or part of an UBA; and can, under appropriate conditions, form a molecular complex comprising the ESB, the UBA and the target molecule.
  • ESBs comprise at least one identity identification portion that allow them to bind to or interact with at least one UBA;
  • the ESB are attached, directly or indirectly, to the UBA.
  • the ESBs bind to the UBAs in a cell or sample, e.g., as part of the assay procedure.
  • the UBAs and/or ESBs comprise a capture region.
  • the capture region is used for the isolation of the UBA/ESB and/or immobilization of the UBA/ESB into a surface.
  • the capture region can be an affinity tag, a bead, a slide or an array.
  • the ESBs comprise common linker moiety, for example, a linker oligo.
  • the common linker oligo is complementary to a common linker oligo in the assayable polymer subunits (APSs) that form the cell origination barcode (COB).
  • Figures 3 and 4 show a schematic representation of one of the embodiments of the invention in which a split pool synthesis approach is used to append the COB to the target molecule/UBA/ESB complex.
  • Figure 3 in Step 1 shows the labeling of cells with UBA-ESB-CL reagents.
  • the UBA provides the specificity for the target molecule to be recognized in a cell.
  • the UBA can be an antibody specific for a surface marker like CD8, or an intracellular epitope like a phospho-epitope on a kinase such as Stat-3.
  • the UBA could be an antisense DNA for a target mRNA in a fixed cell.
  • the UBA is identified with an ESB that has a CL moiety, the latter for later addition of cell specific tagging
  • Step 4 is Step 2 shows the start of the split pool synthesis.
  • the cell population is split into 20 tubes.
  • Cell populations can be split into wells, bead or any suitable surfaces known in the art.
  • an APS unit is added to each tube.
  • the APS binds the UBA/ESB complex via the complementary CLs in the APS and the ESB.
  • each cell in a given tube now has appended to each UBA-ESB pair the same subunit as defined by the tube contents (one of 1-20 APS in this example).
  • Each split population from Step 3 now has a DAPS "tag" polymer subunit added to all DBAAs in that cell.
  • Step 5 the cells from the 20 tubes are pulled into one tube.
  • Step 6 steps 2-4 are repeated to add a second APS to the prior APS.
  • cells in one tube will have a mixture of cells all of which have the APS subunit from the round 2 and one of the 20 APSs used in round 1 in a statistically equal distribution.
  • all polymers are extended by the addition of the same APS.
  • the process is repeated as needed.
  • the epitope/barcode along with linked cell origin signature is read by any methods known in the art including the ones described herein.
  • the number of split pool rounds required is defined by the number of cells in an assay and a statistical estimate of what would give an over-representation of the number of tags that ensure unique COB for each cell. This is given by the following equation:
  • a high certainty of tag uniqueness ensures a high statistical significance for identification of cells/particles as distinct entities.
  • a high certainty of tag uniqeness provides for a high likelihood that two identical COB labels originated from the same cell/particle.
  • the invention provides methods for tagging target molecules within cells indirectly with an ESB.
  • the cell population is treated in a split pool synthesis approach that appends to the epitope specific barcode a second signature that indicates the cell of origin, or a cell origin barcode.
  • UBAs can be antibodies, diabodies, etc. for proteins, or anti-sense DNA tags for RNA and DNA for nucleic acids.
  • ESB can be nucleic acids readable by high throughput sequencing approaches or chemical subunits assayable by mass spectrometry approaches.
  • COBs can be nucleic acids readable by high throughput sequencing approaches or chemical subunits assayable by mass spectrometry approaches.
  • the APS can be specific strands of DNA or DNA-mimics.
  • the APSs can be linked via ligase.
  • Example of enzymes that can be used for ligation include but are not limited to DNA ligase, and RNA ligase such as T4 DNA ligase, T4 RNA ligase, Thermus thermophilus (Tth) ligase, Thermus aquaticus (Taq) DNA ligase, or Pyrococcus furiosus (Pfu) ligase.
  • Chemical ligation can be performed using activating and reducing agents such as carbodiimide, cyanogen bromide (BrCN), imidazole, 1-methylimidazole/carbodiimide/cystamine, N- cyanoimidazole, dithiothreitol (DTT) and ultraviolet light.
  • activating and reducing agents such as carbodiimide, cyanogen bromide (BrCN), imidazole, 1-methylimidazole/carbodiimide/cystamine, N- cyanoimidazole, dithiothreitol (DTT) and ultraviolet light.
  • ligation techniques such as gap-filling ligation, including, without limitation, gap- filling OLA and LCR, bridging oligonucleotide ligation, and correction ligation. Descriptions of these techniques can be found, among other places, in U.S. Pat. No. 5,185,243, published European Patent Applications EP 320308 and EP 439182, and
  • APS subunits can be detected via full sequencing of all DNA tags by any suitable methods known in the art, e.g., Illumina HiSeq 2000, including the sequencing methods described herein.
  • the APS can be small molecules as per combinatorial synthesis procedures or buildable complex molecules of deterministic weights. These subunits can be detected via mass spectrometry
  • the cell-specific information is assembled via a
  • the UBA/ESB/COB complexes are isolated via a capture region as described herein. In some embodiments, the capture region is uses for the
  • the information on UBAs can be amplified prior to the then COB procedure (split pool) with any suitable amplification technique known in the art, including the ones described herein such branch chain or rolling circle approaches.
  • error correction & detection can be encoded into subunits.
  • Error-detection and correction schemes can be either systematic or non-systematic: In a systematic scheme, the transmitter sends the original data, and attaches a fixed number of check bits (or parity data), which are derived from the data bits by some deterministic algorithm. If only error detection is required, a receiver can simply apply the same algorithm to the received data bits and compare its output with the received check bits; if the values do not match, an error has occurred at some point during the transmission.
  • the original message is transformed into an encoded message that has at least as many bits as the original message.
  • Error detection and correction schemes include repetition codes, parity bits, checksums, cyclic redundancy checks (CRCs), cryptographic hash functions, error-correcting codes, automatic repeat request, Hybrid ARQ, error-correcting code, convolutional codes, block codes such as hamming codes, multidimensional parity-check codes, Reed-Solomon codes, turbo codes and low-density parity- check codes (LDPC).
  • the chains at each round are blocked from further addition if polymers did not add. This could be accomplished with DNA as polymer units if each round one uses different overhangs for complementary additions.
  • the epitope/barcode along with linked cell origin signature is read by sequencing using methods known in the art, including the ones described herein.
  • the sensitivity for the sequencing approach assuming that each 100 bp read represents an ESB-COB pair is as follows for target proteins.
  • the protein copy numbers of molecules of interest range from 100 to 100,000. Assuming that one will want to read 100 proteins with the following rough distribution: 1000 copies 20 2 x 10 4 20 2 x 10 4
  • Test 1 one will need to be able to read 2,232,000 sequences per cell then to access all 100 proteins in a cell.
  • a sequencing technique that can do 2 x 10 9 reads such as Illumina HiSeq 2000, this means the approach can read 100 proteins in -1000 cells.
  • we can increase the cell number accessible. Say one therefore normalize and cap the 100,000 copies to a 10,000 copy limit (Test 2), then the total number of reads is 432,000. That is one can read 100 proteins in -5000 cells.
  • RNA copy numbers of molecules of interest range from 5 to 1000 (based on Lewin's Essential Genes numbers); not counting specialized mRNAs for high protein production like actin or Ig. Thus, assuming one wants to read 100 mRNAs with the following rough
  • Test 1 one needs to be able to read 12,300 sequences per cell to access all 100 mRNAs in a cell. Using a sequencing technique that can do 2 x 10 9 reads such as Illumina HiSeq 2000, this means the approach can read 100 mRNAs in -162,000 cells. This is equivalent to a high parameter flow cytometry run. 200 mRNAs with the same distribution could scale linearly to be read on -80,000 cells.
  • the invention provides methods comprising UBA for the analysis of target molecules.
  • the invention provides a UBA population for use in a multiplexed assay. Each UBA in the population is specific for a target molecule. The binding of the target molecules to the UBAs is then detected using ESB-COB pairs. Each ESB-COB pair comprises a unique label code that can be associated to a specific target molecule and the cell of origin as described herein.
  • the detection of the ESB-COB as described below is digital in nature in that one molecule at a time is counted. While fluorescence is used to read the code, the signals are high and the spot is either present of not, thus the digital detection. Using digital detection rather than an analogue fluorescent signal used to quantify signal leads to more accurate quantification. Thus the methods described herein allows for multiplexing to levels beyond currently possible, for more accurate quantification, and possibly higher sensitivity.
  • the UBA and ESB/COB systems of the invention can be used to detect target molecules in any biomolecular sample.
  • the sample may comprise any number of things, including, but not limited to: biological samples, such as cells (including both primary cells and cultured cell lines), cell lysates, or extracts, tissues and tissue extracts; bodily fluids (including, but not limited to, blood, urine, serum, lymph, bile, cerebrospinal fluid, interstitial fluid, aqueous or vitreous humor, colostrum, sputum, amniotic fluid, saliva, anal and vaginal secretions, perspiration and semen, a transudate, an exudate (e.g., fluid obtained from an abscess or any other site of infection or inflammation) or fluid obtained from a joint (e.g., a normal joint or a joint affected by disease such as rheumatoid arthritis, osteoarthritis, gout or septic arthritis)
  • a joint e.g.,
  • the biomolecular samples can be indirectly derived from biological specimens.
  • the biomolecular sample of the invention can be a sample containing isolated proteins from a cell lysate.
  • the biomolecular sample of the invention is generated by subjecting a biological specimen to fractionation, e.g., size fractionation or membrane fractionation.
  • Protein isolation techniques are also well known in the art and kits employing at least some of these techniques are commercially available. Protein isolation techniques typically employ one or more of the following: maceration and cell lysis, including physical, chemical and enzymatic methods; centrifugation; separations by molecular weight, such as size exclusion chromatography and preparative electrophoresis; selective precipitation, for example, salting-in and salting-out procedures; various chromatographic methods; and the like. Detailed descriptions of and relevant protocols for protein purification techniques can be found in, among other places, Marchak et al., Strategies for Protein Purification and Characterization: A Laboratory Course Manual, Cold Spring Harbor Press (1996); Essentials from Cells: A Laboratory Manual, D. Spector and R. Goldman, eds., Cold Spring Harbor Press (2003); R. Simpson, Proteins and Proteomics: A Laboratory Manual, Cold Spring Harbor Press (2003); and D. Liebler,
  • kits can also be used, for example but not limited to, ProteoExtract.TM. Partial Proteome Extraction Kits (P-PEK) and ProteoExtract.TM. Complete Proteome Extraction Kits (C-PEK), available from P-PEK.
  • P-PEK Partial Proteome Extraction Kits
  • C-PEK Complete Proteome Extraction Kits
  • biomolecular samples of the invention may be either native, e.g., not subject to manipulation or treatment, or treated, which can include any number of treatments, including exposure to candidate agents including drugs, genetic engineering (e.g., the addition or deletion of a gene), etc.
  • Biomolecular samples may also include environmental samples, such as those containing bacteria or other organisms, such as diatoms, dinoflagellates, algae, among others, such as in certain marine or earth-based samples.
  • environmental samples such as those containing bacteria or other organisms, such as diatoms, dinoflagellates, algae, among others, such as in certain marine or earth-based samples.
  • COBs/ESB complexes are detected by any means available in the art that is capable of detecting the specific sequences or signals on a given COBs/ESB complex.
  • the information on the UBA, ESB, COB, UBA/ESB complexes, UBA/ESB/COB complexes COB/ESB complexes and/or a combination thereof can be amplified. Amplification can be performed by any means known in the art. In some cases, the information on the UBA, ESB, COB, UBA/ESB complexes, UBA/ESB/COB complexes COB/ESB complexes and/or a combination thereof are amplified by polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • PCR techniques include, but are not limited to, quantitative PCR, quantitative fluorescent PCR (QF-PCR), multiplex fluorescent PCR (MF-PCR), real time PCR (RT-PCR), single cell PCR, restriction fragment length polymorphism PCR (PCR-RFLP), PCR-RFLP/RT-PCR-RFLP, hot start PCR, nested PCR, in situ polonony PCR, in situ rolling circle amplification (RCA), bridge PCR, picotiter PCR and emulsion PCR.
  • QF-PCR quantitative fluorescent PCR
  • MF-PCR multiplex fluorescent PCR
  • RT-PCR real time PCR
  • PCR-RFLP restriction fragment length polymorphism PCR
  • PCR-RFLP PCR-RFLP/RT-PCR-RFLP
  • hot start PCR nested PCR
  • in situ polonony PCR in situ rolling circle amplification
  • RCA in situ rolling circle amplification
  • bridge PCR picotiter PCR and emulsion PCR.
  • LCR ligase chain reaction
  • transcription amplification self- sustained sequence replication
  • selective amplification of target polynucleotide sequences consensus sequence primed polymerase chain reaction (CP-PCR), arbitrarily primed polymerase chain reaction (AP-PCR), degenerate oligonucleotide-primed PCR (DOP-PCR) and nucleic acid based sequence amplification (NABSA).
  • CP-PCR consensus sequence primed polymerase chain reaction
  • AP-PCR arbitrarily primed polymerase chain reaction
  • DOP-PCR degenerate oligonucleotide-primed PCR
  • NABSA nucleic acid based sequence amplification
  • Other amplification methods that can be used herein include those described in U.S. Patent Nos. 5,242,794; 5,494,810; 4,988,617; and 6,582,938.
  • target nucleic acids may be obtained from a single cell.
  • UBA/ESB complexes, UBA/ESB/COB complexes COB/ESB complexes and/or a combination thereof can be pre-amplified prior to the amplification step (e.g., PCR).
  • amplification step e.g., PCR
  • UBA/ESB/COB complexes COB/ESB complexes and/or a combination thereof are quantified.
  • Methods for quantifying nucleic acids include, but are not limited to, gas chromatography, supercritical fluid chromatography, liquid chromatography (including partition chromatography, adsorption chromatography, ion exchange chromatography, size- exclusion chromatography, thin-layer chromatography, and affinity chromatography), electrophoresis (including capillary electrophoresis, capillary zone electrophoresis, capillary isoelectric focusing, capillary electro chromatography, micellar electrokinetic capillary chromatography, isotachophoresis, transient isotachophoresis and capillary gel electrophoresis), comparative genomic hybridization (CGH), microarrays, bead arrays, and high-throughput genotyping such as with the use of molecular inversion probe (MIP).
  • MIP molecular inversion probe
  • Quantification of the UBA, ESB, COB, UBA/ESB complexes, UBA/ESB/COB complexes COB/ESB complexes and/or a combination thereof can be used to determine gene/or allele copy number, gene or exon-level expression, methylation-state analysis, or detect a novel transcript in order to diagnose or condition, i.e. fetal abnormality or cancer.
  • UBA/ESB/COB complexes COB/ESB complexes and/or a combination thereof are sequenced. Sequencing can be accomplished through classic Sanger sequencing methods which are well known in the art. Sequence can also be accomplished using high-throughput systems some of which allow detection of a sequenced nucleotide immediately after or upon its incorporation into a growing strand, i.e., detection of sequence in real time or substantially real time.
  • high throughput sequencing generates at least 1,000, at least 5,000, at least 10,000, at least 20,000, at least 30,000, at least 40,000, at least 50,000, at least 100,000 or at least 500,000 sequence reads per hour; with each read being at least 50, at least 60, at least 70, at least 80, at least 90, at least 100, at least 120 or at least 150 bases per read.
  • high-throughput sequencing involves the use of technology available by Illumina's HiSeq 2000 machine. This machine uses reversible terminator-based sequencing by synthesis chemistry. This machine can do 200 billion DNA reads in eight days.
  • high-throughput sequencing involves the use of technology available by ABI Solid System. This genetic analysis platform that enables massively parallel sequencing of clonally-amplified DNA fragments linked to beads.
  • the sequencing methodology is based on sequential ligation with dye-labeled oligonucleotides.
  • high-throughput sequencing involves the use of technology available by Ion Torrent Personal Genome Machine (PMG).
  • PMG Personal Genome Machine
  • the PGM can do 10 million reads in two hours.
  • high-throughput sequencing involves the use of technology available by Helicos Biosciences Corporation (Cambridge, Massachusetts) such as the Single Molecule Sequencing by Synthesis (SMSS) method. SMSS is unique because it allows for sequencing the entire human genome in up to 24 hours. Finally, SMSS is described in part in US Publication Application Nos. 20060024711; 20060024678; 20060012793;
  • high-throughput sequencing involves the use of technology available by 454 Lifesciences, Inc. (Branford, Connecticut) such as the
  • PicoTiterPlate device which includes a fiber optic plate that transmits chemilluminescent signal generated by the sequencing reaction to be recorded by a CCD camera in the instrument. This use of fiber optics allows for the detection of a minimum of 20 million base pairs in 4.5 hours.
  • high-throughput sequencing is performed using Clonal
  • high-throughput sequencing can take place using
  • AnyDot.chips (Genovoxx, Germany).
  • the AnyDot.chips allow for lOx - 50x enhancement of nucleotide fluorescence signal detection.
  • AnyDot.chips and methods for using them are described in part in International Publication Application Nos. WO 02088382, WO 03020968, WO 03031947, WO 2005044836, PCT/EP 05/05657, PCT/EP 05/05655; and German Patent Application Nos.
  • a polymerase on the target nucleic acid molecule complex is provided in a position suitable to move along the target nucleic acid molecule and extend the oligonucleotide primer at an active site.
  • a plurality of labeled types of nucleotide analogs are provided proximate to the active site, with each distinguishable type of nucleotide analog being complementary to a different nucleotide in the target nucleic acid sequence.
  • the growing nucleic acid strand is extended by using the polymerase to add a nucleotide analog to the nucleic acid strand at the active site, where the nucleotide analog being added is complementary to the nucleotide of the target nucleic acid at the active site.
  • the nucleotide analog added to the oligonucleotide primer as a result of the polymerizing step is identified.
  • the steps of providing labeled nucleotide analogs, polymerizing the growing nucleic acid strand, and identifying the added nucleotide analog are repeated so that the nucleic acid strand is further extended and the sequence of the target nucleic acid is determined.
  • sequence analysis of the UBA, ESB, COB, UBA/ESB complexes, UBA/ESB/COB complexes COB/ESB complexes and/or a combination thereof may include a four-color sequencing by ligation scheme (degenerate ligation), which involves hybridizing an anchor primer to one of four positions. Then an enzymatic ligation reaction of the anchor primer to a population of degenerate nonamers that are labeled with fluorescent dyes is performed. At any given cycle, the population of nonamers that is used is structure such that the identity of one of its positions is correlated with the identity of the fluorophore attached to that nonamer.
  • degenerate ligation degenerate ligation
  • the fluorescent signal allows the inference of the identity of the base.
  • One or more UBA, ESB, COB, UBA/ESB complexes, UBA/ESB/COB complexes COB/ESB complexes and/or a combination thereof complexes can be detected and/or quantified by any method that detects and/or quantifies the presence of the assembled detection complex of interest.
  • Such methods may include radioimmunoassay (RIA) or enzyme linked immunoabsorbance assay (ELISA), immunohistochemistry, immunofluorescent histochemistry with or without confocal microscopy, Raman spectroscopy, X-ray autoradiography, X-ray radiography, luminescence spectrometry, reversed phase assays, homogeneous enzyme immunoassays, and related non-enzymatic techniques, Western blots, whole cell staining, immunoelectron microscopy, nucleic acid amplification, gene array, protein array, mass spectrometry, patch clamp, 2-dimensional gel electrophoresis, differential display gel electrophoresis, microsphere-based multiplex protein assays, label-free cellular assays and flow cytometry, etc.
  • RIA radioimmunoassay
  • ELISA enzyme linked immunoabsorbance assay
  • U.S. Pat. No. 4,568,649 describes ligand detection systems, which employ scintillation counting. These techniques are particularly useful for modified protein parameters. Cell readouts for proteins and other cell determinants can be obtained using fluorescent or otherwise tagged reporter molecules. Microscopy methods are useful for measuring parameters in a morphological context. Flow cytometry methods are useful for measuring intracellular parameters.
  • fluorescent monitoring systems e.g., Cytometric measurement device systems
  • flow cytometric systems are used or systems dedicated to high throughput screening, e.g. 96 well or greater microtiter plates, e.g., Lakowicz, J. R., Principles of
  • Possible sources may include but are not limited to arc lamp, xenon lamp, lasers, light emitting diodes or some combination thereof.
  • the appropriate excitation source is used in conjunction with an appropriate optical detection system, for example an inverted fluorescent microscope, an epi- fluorescent microscope or a confocal microscope.
  • an appropriate optical detection system for example an inverted fluorescent microscope, an epi- fluorescent microscope or a confocal microscope.
  • a microscope is used that can allow for detection with enough spatial resolution to determine the sequence of the spots on the COB/ESB complexes. If for example, the COB/ESB complexes are labeled with three different colors, Alexa 488, Cy3 and Alexa 647 (labeled 1, 2 and 3, respectively).
  • Colors 1, 2 and 3 are each acquired in different channels and the first and second registers, which can be seen as rows of spots, are shifted up by several pixels to be able to show each register individually. Examples of methods for detection of multiple colors that can be used in the methods of the invention are described in US patent 7,473,767, US patent publication no. 2007/0166708, US application number 11/645,270, and PCT application no US06/049274, incorporated by reference herein in its entirety.
  • Fluorescence in a sample can be measured using a fluorimeter.
  • Other methods of detecting fluorescence may also be used, e.g., Quantum dot methods (see, e.g., Goldman et al, J. Am. Chem. Soc. (2002) 124:6378-82; Pathak et al. J. Am. Chem. Soc. (2001) 123:4103-4; and Remade et al, Proc. Natl. Sci. USA (2000) 18:553-8, each expressly incorporated herein by reference) as well as confocal microscopy.
  • a FACS cell sorter e.g. a FACSVantageTM, LSRII, or
  • Canto Cell Sorter Becton Dickinson Immunocytometry Systems, San Jose, Calif.
  • Canto Cell Sorter is used to sort and collect cells based on the presence or absence of an COB/ESB complex. By imparting an electromagnetic charge to droplets containing the positive cells, the cells can be separated from other cells. The positively selected cells can then be harvested in sterile collection vessels.
  • positive cells can be sorted using magnetic separation of cells based on the presence of a COB/ESB complex.
  • cells to be positively selected are first contacted with a COB/ESB complex comprising retrievable particles (e.g., magnetically responsive particles).
  • the cell can then be physically separated from non- positive or non-labeled cells, for example, using a magnetic field.
  • the positive or labeled cells can be retained in a container using a magnetic field while the negative cells are removed.
  • one or more cells are contained in a well of a 96 well plate or other commercially available multi-well plate.
  • the reaction mixture or cells are in a cytometric measurement device.
  • Other multi-well plates useful in the present invention include, but are not limited to 384 well plates and 1536 well plates. Still other vessels for containing the reaction mixture or cells and useful in the present invention will be apparent to the skilled artisan.
  • the abundance of a COB/ESB complex is measured using
  • ICP-MS Inductively Coupled Plasma Mass Spectrometer
  • the 'flow cytometer' is a microfluidic device where the cell measurements, or some of the measurements of the cell's contents, are carried out in channels devised to direct cells past detection devices in parallel sets of multiple channels. See U. S. Patent Nos. 7,378,280; 7,294,503; 7,294,298; and 6,830,936.
  • the cells, or some portion of their contents are sonically encapsulated within individual droplets of liquid and interrogated with detection devices designed to measure each individual droplet' s characteristics and the materials within such droplets.
  • the software program modules allow creation, modification, and running of methods.
  • the system diagnostic modules allow instrument alignment, correct connections, and motor operations.
  • Customized tools, labware, and liquid, particle, cell and organism transfer patterns allow different applications to be performed.
  • Databases allow method and parameter storage. Robotic and computer interfaces allow communication between instruments.
  • the methods of the invention include the use of liquid handling components.
  • the liquid handling systems can include robotic systems comprising any number of components.
  • any or all of the steps outlined herein may be automated; thus, for example, the systems may be completely or partially automated.
  • Fully robotic or microfluidic systems include automated liquid-, particle-, cell- and organism-handling including high throughput pipetting to perform all steps of screening applications.
  • This includes liquid, particle, cell, and organism manipulations such as aspiration, dispensing, mixing, diluting, washing, accurate volumetric transfers; retrieving, and discarding of pipet tips; and repetitive pipetting of identical volumes for multiple deliveries from a single sample aspiration.
  • These manipulations are cross-contamination-free liquid, particle, cell, and organism transfers.
  • This instrument performs automated replication of microplate samples to filters, membranes, and/or daughter plates, high-density transfers, full-plate serial dilutions, and high capacity operation.
  • chemically derivatized particles, plates, cartridges, tubes, magnetic particles, or other solid phase matrix with specificity to the assay components are used.
  • the binding surfaces of microplates, tubes or any solid phase matrices include non-polar surfaces, highly polar surfaces, modified dextran coating to promote covalent binding, antibody coating, affinity media to bind fusion proteins or peptides, surface-fixed proteins such as recombinant protein A or G, nucleotide resins or coatings, and other affinity matrix are useful in this invention.
  • platforms for multi-well plates, multi-tubes, holders, cartridges, minitubes, sonic levitation and encapsulation, deep-well plates, microfuge tubes, cryovials, square well plates, filters, chips, microchannel chips, microfluidics chips, optic fibers, beads, and other solid-phase matrices or platform with various volumes are accommodated on an upgradeable modular platform for additional capacity.
  • This modular platform includes a variable speed orbital shaker, and multi-position work decks for source samples, sample and reagent dilution, assay plates, sample and reagent reservoirs, pipette tips, and an active wash station.
  • the methods of the invention include the use of a plate reader.
  • thermocycler and thermoregulating systems are used for stabilizing the temperature of heat exchangers such as controlled blocks or platforms to provide accurate temperature control of incubating samples from 0° C. to 100° C.
  • interchangeable pipet heads with single or multiple magnetic probes, affinity probes, or pipetters robotically manipulate the liquid, particles, cells, and organisms.
  • Multi-well or multi-tube magnetic separators or platforms manipulate liquid, particles, cells, and organisms in single or multiple sample formats.
  • the instrumentation will include a detector, which can be a wide variety of different detectors, depending on the labels and assay.
  • useful detectors include a microscope(s) with multiple channels of fluorescence; plate readers to provide fluorescent, ultraviolet and visible spectrophotometric detection with single and dual wavelength endpoint and kinetics capability, fluorescence resonance energy transfer (FRET), luminescence, quenching, two-photon excitation, and intensity redistribution; CCD cameras to capture and transform data and images into quantifiable formats; and a computer workstation.
  • the robotic apparatus includes a central processing unit which communicates with a memory and a set of input/output devices (e.g., keyboard, mouse, monitor, printer, etc.) through a bus. Again, as outlined below, this may be in addition to or in place of the CPU for the multiplexing devices of the invention.
  • a central processing unit which communicates with a memory and a set of input/output devices (e.g., keyboard, mouse, monitor, printer, etc.) through a bus.
  • input/output devices e.g., keyboard, mouse, monitor, printer, etc.
  • this may be in addition to or in place of the CPU for the multiplexing devices of the invention.
  • the general interaction between a central processing unit, a memory, input/output devices, and a bus is known in the art. Thus, a variety of different procedures, depending on the experiments to be run, are stored in the CPU memory.
  • robotic fluid handling systems can utilize any number of different reagents, including buffers, reagents, samples, washes, assay components such as label probes, etc.
  • compositions and methods of the invention can be used for diagnostic, prognostic, therapeutic, patient stratification, drug development, treatment selection, and screening purposes.
  • the present invention provides the advantage that many different target molecules can be analyzed at one time from a single biomolecular sample using the methods of the invention. This allows, for example, for several diagnostic tests to be performed on one sample.
  • composition and methods of the invention can be used in proteomics.
  • the methods described herein will typically provide an answer rapidly which is very desirable for this application.
  • the methods and composition described herein can be used in the process of finding biomarkers that may be used for diagnostics or prognostics and as indicators of health and disease.
  • the methods and composition described herein can be used to screen for drugs, e.g., drug development, selection of treatment, determination of treatment efficacy and/or identify targets for pharmaceutical development.
  • the ability to test protein expression on screening assays involving drugs is very important because proteins are the final gene product in the body.
  • the methods and compositions described herein will measure both protein and gene expression simultaneously which will provide the most information regarding the particular screening being performed.
  • composition and methods of the invention can be used in gene expression analysis.
  • the methods described herein discriminate between nucleotide sequences.
  • the difference between the target nucleotide sequences can be, for example, a single nucleic acid base difference, a nucleic acid deletion, a nucleic acid insertion, or rearrangement. Such sequence differences involving more than one base can also be detected.
  • the UBAs e.g., oligonucleotide probe, have substantially the same length so that they hybridize to target nucleotide sequences at substantially similar hybridization conditions. As a result, the process of the present invention is able to detect infectious diseases, genetic diseases, and cancer.
  • nucleic acids It is also useful in environmental monitoring, forensics, and food science.
  • genetic analyses that can be performed on nucleic acids include e-g., SNP detection, STR detection, RNA expression analysis, promoter methylation, gene expression, virus detection, viral subtyping and drug resistance.
  • the present methods can be applied to the analysis of biomolecular samples obtained or derived from a patient so as to determine whether a diseased cell type is present in the sample, the stage of the disease, the prognosis for the patient, the ability to the patient to respond to a particular treatment, or the best treatment for the patient.
  • the present methods can also be applied to identify biomarkers for a particular disease.
  • the methods described herein are used in the diagnosis of a condition.
  • diagnosis or “diagnosis” of a condition includes predicting or diagnosing the condition, determining predisposition to the condition, monitoring treatment of the condition, diagnosing a therapeutic response of the disease, and prognosis of the condition, condition progression, and response to particular treatment of the condition.
  • a blood sample can be assayed according to any of the methods described herein to determine the presence and/or quantity of markers of a disease or malignant cell type in the sample, thereby diagnosing or staging the a disease or a cancer.
  • the methods and composition described herein are used for the diagnosis and prognosis of a condition.
  • Immunologic diseases and disorders include allergic diseases and disorders, disorders of immune function, and autoimmune diseases and conditions.
  • Allergic diseases and disorders include but are not limited to allergic rhinitis, allergic conjunctivitis, allergic asthma, atopic eczema, atopic dermatitis, and food allergy.
  • Immunodeficiencies include but are not limited to severe combined immunodeficiency (SCID), hypereosinophilic syndrome, chronic granulomatous disease, leukocyte adhesion deficiency I and II, hyper IgE syndrome, Chediak Higashi, neutrophilias, neutropenias, aplasias,
  • SCID severe combined immunodeficiency
  • Autoimmune and immune dysregulation disorders include but are not limited to rheumatoid arthritis, diabetes, systemic lupus erythematosus, Graves' disease, Graves ophthalmopathy, Crohn's disease, multiple sclerosis, psoriasis, systemic sclerosis, goiter and struma lymphomatosa (Hashimoto's thyroiditis, lymphadenoid goiter), alopecia aerata, autoimmune myocarditis, lichen sclerosis, autoimmune uveitis, Addison's disease, atrophic gastritis, myasthenia gravis, idiopathic thrombocytopenic purpura, hemolytic anemia, primary biliary cirrhosis, Wegener's granulomatosis,
  • Proliferative diseases and disorders that may be evaluated by the methods of the invention include, but are not limited to, hemangiomatosis in newborns; secondary progressive multiple sclerosis; chronic progressive myelodegenerative disease; neurofibromatosis;
  • ganglioneuromatosis ganglioneuromatosis; keloid formation; Paget's Disease of the bone; fibrocystic disease (e.g., of the breast or uterus); sarcoidosis; Peronies and Duputren's fibrosis, cirrhosis, atherosclerosis and vascular restenosis.
  • Malignant diseases and disorders that may be evaluated by the methods of the invention include both hematologic malignancies and solid tumors.
  • Hematologic malignancies are especially amenable to the methods of the invention when the sample is a blood sample, because such malignancies involve changes in blood-borne cells.
  • Such malignancies include non-Hodgkin's lymphoma, Hodgkin's lymphoma, non-B cell lymphomas, and other lymphomas, acute or chronic leukemias, polycythemias, thrombocythemias, multiple myeloma, myelodysplasia disorders, myeloproliferative disorders, myelofibroses, atypical immune lymphoproliferations and plasma cell disorders.
  • Plasma cell disorders that may be evaluated by the methods of the invention include multiple myeloma, amyloidosis and Waldenstrom's macroglobulinemia.
  • Example of solid tumors include, but are not limited to, colon cancer, breast cancer, lung cancer, prostate cancer, brain tumors, central nervous system tumors, bladder tumors, melanomas, liver cancer, osteosarcoma and other bone cancers, testicular and ovarian carcinomas, head and neck tumors, and cervical neoplasms.
  • detectable genetic diseases include: 21 hydroxylase deficiency, cystic fibrosis, Fragile X Syndrome, Turner Syndrome, Duchenne Muscular Dystrophy, Down Syndrome or other trisomies, heart disease, single gene diseases, HLA typing, phenylketonuria, sickle cell anemia, Tay-Sachs Disease, thalassemia, Klinefelter Syndrome, Huntington Disease, autoimmune diseases, lipidosis, obesity defects, hemophilia, inborn errors of metabolism, and diabetes.
  • the methods described herein can be used to diagnose pathogen infections, for example infections by intracellular bacteria and viruses, by determining the presence and/or quantity of markers of bacterium or virus, respectively, in the sample.
  • infectious diseases can be detected by the process of the present invention. Typically, these are caused by bacterial, viral, parasite, and fungal infectious agents. The resistance of various infectious agents to drugs can also be determined using the present invention.
  • Bacterial infectious agents which can be detected by the present invention include Escherichia coli, Salmonella, Shigella, KlESBiella, Pseudomonas, Listeria
  • Fungal infectious agents which can be detected by the present invention include
  • Cryptococcus neoformans Blastomyces dermatitidis, Histoplasma capsulatum, Coccidioides immitis, Paracoccidioides brasiliensis, Candida albicans, Aspergillus fumigautus, Phycomycetes (Rhizopus), Sporothrix schenckii, Chromomycosis, and Maduromycosis.
  • Viral infectious agents which can be detected by the present invention include human immunodeficiency virus, human T-cell lymphocytotrophic virus, hepatitis viruses (e.g., Hepatitis B Virus and Hepatitis C Virus), Epstein-Barr Virus, cytomegalovirus, human papillomaviruses, orthomyxo viruses, paramyxo viruses, adenoviruses, corona viruses, rhabdo viruses, polio viruses, toga viruses, bunya viruses, arena viruses, rubella viruses, and reo viruses.
  • human immunodeficiency virus e.g., human T-cell lymphocytotrophic virus
  • hepatitis viruses e.g., Hepatitis B Virus and Hepatitis C Virus
  • Epstein-Barr Virus Epstein-Barr Virus
  • cytomegalovirus human papillomaviruses
  • orthomyxo viruses paramyxo viruses
  • corona viruses coron
  • Parasitic agents which can be detected by the present invention include
  • Plasmodium falciparum Plasmodium malaria
  • Plasmodium vivax Plasmodium ovale
  • Onchoverva volvulus Leishmania, Trypanosoma spp., Schistosoma spp., Entamoeba histolytica, Cryptosporidum, Giardia spp., Trichimonas spp., Balatidium coli, Wuchereria bancrofti, Toxoplasma spp., Enterobius vermicularis, Ascaris lumbricoides, Trichuris trichiura,
  • the present invention is also useful for detection of drug resistance by infectious agents.
  • infectious agents for example, vancomycin-resistant Enterococcus faecium, methicillin-resistant
  • Staphylococcus aureus penicillin-resistant Streptococcus pneumoniae, multi-drug resistant Mycobacterium tuberculosis, and AZT -resistant human immunodeficiency virus can all be identified with the present invention
  • the target molecules detected using the compositions and methods of the invention can be either patient markers (such as a cancer marker) or markers of infection with a foreign agent, such as bacterial or viral markers.
  • compositions and methods of the invention can be used to quantify target molecule whose abundance is indicative of a biological state or disease condition, for example, blood markers that are upregulated or downregulated as a result of a disease state.
  • the methods and compositions of the present invention can be used for cytokine detection.
  • the low sensitivity of the methods described herein would be helpful for early detection of cytokines, e.g., as biomarkers of a condition, diagnosis or prognosis of a disease such as cancer, and the identification of subclinical conditions.
  • kits comprising one or more components of the invention.
  • the kits can comprise, for example, one or more UBAs, one or more ESBs, and/or one or more APSs.
  • the kits can be used for any purpose apparent to those of skill in the art, including those described above.
  • the present invention also provides kits useful for the extension and selective immobilization of COBs, UBAs, ESBs and/or a combination thereof.
  • the kits can comprise a substrate for immobilization and one or more binding partners to facilitate extension or immobilization of a COB, UBA, ESB and/or a combination thereof.
  • the binding partners could, in certain embodiments, comprise a moiety useful for extension of the COB, UBA, ESB and/or a combination thereof, in an appropriate force. In certain embodiments, the binding partners could facilitate immobilization or selective immobilization of the COB, UBA, ESB and/or a combination thereof, to the surface. In further embodiments, the kits could comprise a device capable of extending the COB, UBA, ESB and/or a combination thereof.
  • kits can contain a population of COBs, APSs, UBAs, ESBs and/or a combination thereof as described herein.
  • kits can contain pre-labeled APSs, or unlabeled APSs with one or more components for labeling the APSs.
  • the ESBs and/or APSs provided in a kit may or may not have UBAs pre-attached.
  • the UBAs are provided in the kit unattached to the ESBs and/or APSs.
  • kits can comprise other reagents such as linker oligos and bridging oligos.
  • kits can separate the UBAs into different premixes.
  • kits can include other reagents as well, for example, buffers for performing hybridization reactions, linkers, restriction endonucleases, and DNA I ligases.
  • kits also will include instructions for using the components of the kit, and/or for making and/or using the APSs, COBs, UBAs, and/or ESBs.
  • Oligonucleotides can be synthesized according to standard techniques known in the art. For instance, oligonucleotides can be synthesized on a 394 A DNA Synthesizer (Applied to the art).
  • the oligonucleotides are purified by ethanol precipitation after overnight deprotection at 55° C.
  • the primer-specific portions of the oligonucleotides used for PCR amplification are purified by polyacrylamide gel electrophoresis on 10% acrylamide/7M urea gels.
  • Oligonucleotides are visualized after electrophoresis by UV shadowing against a lightening screen and excised from the gel (Applied Biosystems Inc., 1992). They are then eluted overnight at 64° C. in TNE (i.e. Tris-sodium EDTA) buffer (100 mM Tris/HCl pH 8.0 containing 500 mM NaCl and 5 mM EDTA) and recovered from the eluate using Sep Pak cartridges (Millipore Corp, Milford, Mass.) following the manufacturer's instructions. [00259] Oligonucleotides are resuspended in 100 ⁇ TE (i.e. 10 mM Tri-HCl pH 8.0 containing 1 mM EDTA). Typical concentrations of these original oligonucleotide solutions are about 1 ⁇ g/ ⁇ l or approximately 74 pm/ ⁇ .
  • the oligonucleotides are phosphorylated with T4 polynucleotide kinase at the 5 '-end. Aliquots of the oligonucleotides equivalent to 200 pm are combined with 10 ⁇ of lOx kinase buffer (500 mM Tris/HCl pH 8.0, 100 mM MgCl 2 ), 10 ⁇ of 10 mM ATP, 20 U T4 kinase, and sufficient water-ME to give a final volume of 100 ⁇ .
  • lOx kinase buffer 500 mM Tris/HCl pH 8.0, 100 mM MgCl 2
  • 10 ⁇ of 10 mM ATP 10 ⁇ of 10 mM ATP
  • 20 U T4 kinase 20 U T4 kinase
  • Phosphorylation is carried out at 37° C. for 30 min followed by incubation for 10 min at 85° C to inactivate the T4 enzyme.
  • the solutions of the oligonucleotides are adjusted to convenient concentrations.
  • the kinased oligonucleotide solution is diluted fourfold in water to yield a concentration of 1000 frn/ ⁇ .
  • a solution of the oligonucleotides is made by combining volumes of the oligonucleotides equivalent to 200 pm with sufficient water to give a final volume of 400 ⁇ . This created a solution 1000 fm/ ⁇ in each of the oligonucleotides. Aliquots (20 ⁇ ) of the kinased and unkinased oligonucleotides are frozen for subsequent use.
  • Oligonucleotides are synthesized as described in El-Sagheer et al. (PNAS, 108:28, 11338-11343, 2011). Briefly, standard DNA phosphoramidites, solid supports and additional reagents are purchased from Link Technologies and Applied Biosystems. Oligonucleotides are synthesized on an Applied Biosystems 394 automated DNA/ RNA synthesizer using a standard 0.2 or 1.0 //mole phosphoramidite cycle of acid-catalyzed detritylation, coupling, capping, and iodine oxidation.
  • oligonucleotides from the solid support and deprotection is achieved by exposure to concentrated aqueous ammonia solution for 60 min at room temperature followed by heating in a sealed tube for 5 hr at 55 °C.
  • the oligonucleotides are purified by reversed-phase HPLC on a Gilson system using an XBridgeTM BEH300 Prep C18 10 ⁇ 10x250 mm column (Waters) with a gradient of acetonitrile in ammonium acetate (0% to 50% buffer B over 30 min, flow rate 4 mL/min), buffer A: 0.1 M ammonium acetate, pH 7.0, buffer B: 0.1 M ammonium acetate, pH 7.0, with 50% acetonitrile.
  • 3 '-alkyne oligonucleotides are synthesized using the 3'-propargylthymidine phosphoramidite monomer 2c and assembling the required sequence in the 5' to 3 '-direction using the 3'-0-(4,4'-dimethoxytrityl)deoxyribonucleoside-5'- phosphor-amidites of A, G, C and T (reverse phosphoramidites, Link Technologies) or by the attachment of 5'-0-(4,4'-dimethoxytrityl)-3'-O-propargyl-5-methyl-deoxycytidine to solid support (33 ⁇ /g loading, AM polystyrene, Applied Biosystems) according to El-Sagheer
  • oligonucleotides are assembled on the 0.2 or 1.0 ⁇ scale (trityl-off) as described in the general method (above) with normal 5'-HO-dC, 5'- HO-dT (or with 5'-iodo-dT using the commercially available 5'-iodo dT monomer from Glen Research).
  • the protected oligomers attached to the synthesis column are treated with a 0.5 M solution of methyltriphenoxyphosphonium iodide in DMF (1.0 mL), which is periodically passed through the column via two 1 mL syringes over 15 min at room temperature. The column is then washed several times with dry DMF.
  • Oligonucleotide APSs are annealed to a template and kept overnight at 4° C.
  • a solution of Cu 1 click catalyst is prepared from tm-hydroxypropyltriazole ligand as described in Chan et al. (Chan TR, Hilgraf R, Sharpless KB, & Fokin VV (2004) Polytriazoles as coppers- stabilizing ligands in catalysis. Org. Lett.
  • COBs are synthesized attached to beads. 4 different methods are used for the assembly of APSs into COBs:
  • Aminomethyl macroporous polystyrene (MPPS) beads are labeled with ten different CL oligonucleotides, each with an optional first amplification primer complementary region, one of 10 different ESB sequences and a common annealing region. Six rounds of split pool synthesis are performed. In each round, beads are split into 20 different containers. A different oligonucleotide APS is added to each container, totaling 20 different APSs. Each APS in a given round further comprises a unique subcode sequence that is different from the rest of the APSs in that round.
  • each APS comprises an annealing region 1 that is
  • each APS comprises an annealing region complementary to the available annealing region of the APS from the previous round on one end and an additional annealing region on the other end.
  • the added APS hybridizes to the APS added in the previous round along the complementary annealing region.
  • the last subunit optionally comprises a second amplification primer complimentary region for hybridization of PCR or sequencing primers.
  • a CL, or one or more APSs further comprise a random tag region, which acts as a molecular counter as described supra, allowing for subsequent normalization of the detected COBs.
  • the beads are pooled and divided into new 20 pools initiating the subsequent round.
  • a new set of 20 APSs are added in each round with a pair of round specific annealing region as described above.
  • the hybridized APSs on the beads are patched together using a polymerase/ligase.
  • the COBs are optionally PCR amplified for sequencing using primers targeting the amplification primer complementary regions on the CL and the last APS subunit
  • Aminomethyl macroporous polystyrene (MPPS) beads are labeled with ten different CL oligonucleotides, each with an optional first amplification primer complementary region, a one of 10 different ESB sequences and a common annealing region. Six rounds of split pool synthesis are performed. In each round, beads are split into 20 different containers. A different oligonucleotide APS is added to each container, totaling 20 different APSs. Each APS in a given round further comprises a unique subcode sequence that is different from the rest of the APSs in that round.
  • an annealing primer is also added.
  • the annealing primer has a complementary region to the CL oligonucleotide and a complementary region to the APS.
  • the annealing primer hybridizes to both, stitching them together.
  • the annealing primer has a complementary region to the APS added during the previous round and a complementary region to the APS being added in the current round.
  • the annealing primer hybridizes to APSs from subsequent rounds stitching them together.
  • the complementary regions of the annealing primer are specific to each round allowing efficient hybridization of subunits only from the previous and current rounds. Accordingly, the annealing primer does not hybridize to subunits of earlier rounds, which would not have complementary regions to the annealing primer of a current round, thus blocking the further synthesis of COBs missing subunits of particular rounds.
  • the last subunit optionally comprises a second amplification primer
  • a CL, or one or more APSs further comprise a random tag region, which acts as a molecular counter as described supra, allowing for subsequent normalization of the detected COBs.
  • the beads are pooled and divided into new 20 pools initiating the subsequent round.
  • a new set of 20 APSs are added in each round with a pair of round specific annealing region as described above.
  • the hybridized APSs on the beads are permanently stitched together using a polymerase/ligase or using Click chemistry as described in Example 2.
  • the COBs are optionally PCR amplified for sequencing using primers targeting the amplification primer complementary regions on the CL and the last APS subunit.
  • Aminomethyl macroporous polystyrene (MPPS) beads are labeled with ten different CL oligonucleotides, each with an optional first amplification primer complementary region, a one of 10 different ESB sequences and a common annealing region. Six rounds of split pool synthesis are performed. In each round, beads are split into 20 different containers. A different oligonucleotide APS is added to each container, totaling 20 different APSs. Each APS in a given round further comprises a unique subcode sequence that is different from the rest of the APSs in that round.
  • an annealing primer is also added.
  • the annealing primer has a first complementary region to the CL oligonucleotide and a second complementary region to the APS being added in the current round.
  • the annealing primer hybridizes to both, stitching them together.
  • the annealing primer has a first complementary region to the APS added during the previous round and a second complementary region to the APS being added in the current round.
  • the annealing primer hybridizes to APSs from subsequent rounds stitching them together.
  • the first complementary regions are specific to each round allowing efficient hybridization to subunits only from the previous round. Accordingly, the annealing primer does not hybridize to subunits of earlier rounds, which would not have complementary regions to the annealing primer of a current round, thus blocking the further synthesis of COBs missing subunits of particular rounds.
  • the last subunit optionally comprises a second amplification primer
  • a CL, or one or more APSs further comprise a random tag region, which acts as a molecular counter as described supra, allowing for subsequent normalization of the detected COBs.
  • the beads are pooled and divided into new 20 pools initiating the subsequent round.
  • a new set of 20 APSs are added in each round with a pair of round specific annealing region as described above.
  • the hybridized APSs on the beads are permanently stitched together using a polymerase/ligase or using Click chemistry as described in Example 2.
  • the COBs are optionally PCR amplified for sequencing using primers targeting the amplification primer complementary regions on the CL and the last APS subunit.
  • Aminomethyl macroporous polystyrene (MPPS) beads are labeled with ten different CL oligonucleotides, each with an optional first amplification primer complementary region, one of 10 different ESB sequences, six pairs of APS-specific loop annealing regions and an optional second amplification primer complementary region. Six rounds of split pool synthesis are performed. In each round, beads are split into 20 different containers. A different oligonucleotide APS is added to each container, totaling 20 different APSs. Each APS in a given round further comprises a unique subcode sequence that is different from the rest of the APSs in that round.
  • the APSs are designed to hybridize to the CL in a loop geometry, hybridizing on each end to the CL along the loop annealing regions specific to the round. The hybridization populates the APSs along the CL, which are then linked together.
  • the APSs are designed such that they do not efficiently hybridize to the CL along the loop annealing regions specific to other rounds. Consequently, if an APS from a particular round is missing, the APSs may not be linked together successfully, depending on the linking process.
  • a COB is synthesized with a missing APS, the location of which is flanked by a pair of loop annealing regions. The resulting COB can then be analyzed accordingly and can either be discarded or the retrieved information can be alternatively processed.
  • a CL, or one or more APSs further comprise a random tag region, which acts as a molecular counter as described supra, allowing for subsequent normalization of the detected COBs.
  • the beads are pooled and divided into new 20 pools initiating the subsequent round.
  • a new set of 20 APSs are added in each round with a pair of round specific annealing region as described above.
  • the hybridized APSs on the beads are permanently stitched together using a polymerase/ligase or using Click chemistry as described in Example 1.
  • the COBs are optionally PCR amplified for sequencing using primers targeting the amplification primer complementary regions on the CL.
  • Aminomethyl macroporous polystyrene (MPPS) beads are labeled with ten different CL oligonucleotides, each with a pair of loop annealing region specific for one of the 10 different loop ESB sequences, and six pairs of APS-specific loop annealing regions. All 10 loop ESB sequences are added to anneal to the loop ESB specific portion of the CL in a loop geometry. Loop ESB sequences are designed to minimize non-specific annealing to the remainder of the loop ESB specific regions of the CLs.
  • Loop ESB sequences comprise an optional first amplification primer complementary region, an ESB sequence, and a pair of annealing regions sufficiently complementary to the loop ESB-specific loop annealing regions in the CL.
  • Six rounds of split pool synthesis are performed. In each round, beads are split into 20 different containers. A different oligonucleotide APS is added to each container, totaling 20 different APSs. Each APS in a given round further comprises a unique subcode sequence that is different from the rest of the APSs in that round. The APSs in the final round optionally further comprise a second amplification primer complementary region.
  • the APSs are designed to hybridize to the CL in a loop geometry, hybridizing on each end to the CL along the loop annealing regions specific to the round. The hybridization populates the APSs along the CL, which are then linked together.
  • the APSs are designed such that they do not efficiently hybridize to the CL along the loop annealing regions specific to other rounds. Consequently, if an APS from a particular round is missing, the APSs may not be linked together successfully, depending on the linking process.
  • a COB is synthesized with a missing APS, the location of which is flanked by a pair of loop annealing regions. The resulting COB can then be analyzed accordingly and can either be discarded or the retrieved information can be alternatively processed.
  • a CL, or one or more APSs further comprise a random tag region, which acts as a molecular counter as described supra, allowing for subsequent normalization of the detected COBs.
  • the beads are pooled and divided into new 20 pools initiating the subsequent round.
  • a new set of 20 APSs are added in each round with a pair of round specific annealing region as described above.
  • the hybridized APSs on the beads are permanently stitched together using a using Click chemistry as described in Example 2.
  • the COBs are optionally PCR amplified for sequencing using primers targeting the amplification primer complementary regions on the CL and the last APS subunit.
  • sequences 3 are sequenced by Illumina's HiSeq 2000 machine.
  • the resulting sequences comprise at least one of 10 different ESB sequences, a random tag region, and a combination of 6 subcodes originating from APSs added to that particular bead during the 6 rounds of split pool synthesis.
  • ESB-linked COBs are synthesized using any of the methods in Example 3.
  • the resulting sequences comprise a T7 promoter an SP6 start site, a start codon, an ESB, a COB and optionally a region encoding a His(6) tag ( Figure 11).
  • the T7 promoter and SP6 start site can be incorporated into the sequence linked to the ESB, using the same method that is used to incorporated the ESB. Alternatively, these sequences can be incorporated withing the last APS.
  • a His(6) tag encoding region can be incorporated linked to the final APS or to the ESB.
  • the assembled ESB-linked COBs are transcribed and translated into peptide sequences using the ExpresswayTM Maxi Cell-Free E. coli Expression System (Invitrogen).
  • the peptide sequences are isolated using affinity chromatography and/or HPLC prior to being sequenced using a tandem mass spectrometer.
  • CD4 are conjugated with amine modified CL oligonucleotides described in Example 3.
  • the singly-labeled antibodies are isolated using affinity chromatography using complementary oligonucleotides targeting a sequence in CL oligonucleotides and the number of labels on each antibody is verified using mass spectrometry.
  • a cell suspension of 10 7 cells are incubated with the combination of the antibodies under suitable conditions followed by 6 rounds of split-pool synthesis.
  • the resulting ESB-linked COBs are detected as described in Example 3 or Example 4.
  • the detected signals related to COB-linked ESBs are quantified for each COB combination.
  • Co- expression of each of the CD1, CD3, CD8, and CD4 antigens on the cells are plotted pairwise. Principle component analysis is used to identify the strongest correlations in expression profiles.
  • Methanol is cooled to -20 °C.
  • a cell culture comprising 10 7 HeLa cells is grown using suitable tissue culture conditions known in the art. The growth medium is removed by aspiration. The cells are immediately fixed and permeabilized by adding 50 mL cold methanol. The cells are allowed to incubate at ambient temperature for 10 minutes with gentle shaking. Methanol is carefully remived by aspiration. The cells are rinsed with 100 mL of IX PBS, three times.
  • the cells are blocked with 150 ml of 0.1% Casein solution in 0.2% PBS for 1.5 hrs at room temperature by gentle shaking.
  • Rabbit anti-cleaved caspase-3, rabbit anti phos-p38, rabbit anti-phos-ERK2, mouse anti-ERK2, and mouse anti-P-tubulin (clone AA2) are CL- conjugated as described in Example 5.
  • Cells are incubated with CL-conjugated antibodies overnight at 4 °C, gently shaking.
  • the cells are washed 5 times with IX PBS + 0.1% Tween-20 for 5 minutes at room temperature, followed by 6 rounds of split-pool synthesis.
  • Example 4 The detected signals related to COB-linked ESBs are quantified for each COB combination. Co-expression of each of the Phospho-p53, ER 1 in the cells are plotted pairwise. Principle component analysis is used to identify the strongest correlations in expression profiles.
PCT/US2012/023411 2011-01-31 2012-01-31 Methods of identifying multiple epitopes in cells WO2012106385A2 (en)

Priority Applications (27)

Application Number Priority Date Filing Date Title
JP2013551435A JP6069224B2 (ja) 2011-01-31 2012-01-31 細胞において複数のエピトープを同定する方法
CN201280016828.5A CN103703143B (zh) 2011-01-31 2012-01-31 鉴定细胞中的多个表位的方法
US13/981,711 US10144950B2 (en) 2011-01-31 2012-01-31 Methods of identifying multiple epitopes in cells
EP12741485.2A EP2670863B1 (en) 2011-01-31 2012-01-31 Methods of identifying multiple epitopes in cells
ES12741485.2T ES2687761T3 (es) 2011-01-31 2012-01-31 Métodos de identificación de múltiples epítopos en células
US15/597,917 US10626442B2 (en) 2011-01-31 2017-05-17 Methods of identifying multiple epitopes in cells
US16/147,250 US10982256B2 (en) 2011-01-31 2018-09-28 Methods of identifying multiple epitopes in cells
US16/163,486 US10982271B2 (en) 2011-01-31 2018-10-17 Methods of identifying multiple epitopes in cells
US16/250,974 US10995362B2 (en) 2011-01-31 2019-01-17 Methods of identifying multiple epitopes in cells
US16/691,493 US20200157603A1 (en) 2011-01-31 2019-11-21 Methods of identifying multiple epitopes in cells
US16/795,203 US20200208197A1 (en) 2011-01-31 2020-02-19 Methods of identifying multiple epitopes in cells
US16/808,100 US11560585B2 (en) 2011-01-31 2020-03-03 Methods of identifying multiple epitopes in cells
US17/071,917 US20210040538A1 (en) 2011-01-31 2020-10-15 Methods of identifying multiple epitopes in cells
US17/204,325 US20220056515A1 (en) 2011-01-31 2021-03-17 Methods of identifying multiple epitopes in cells
US17/870,697 US11566278B2 (en) 2011-01-31 2022-07-21 Methods of identifying multiple epitopes in cells
US17/870,641 US11512341B1 (en) 2011-01-31 2022-07-21 Methods of identifying multiple epitopes in cells
US17/951,003 US11634752B2 (en) 2011-01-31 2022-09-22 Kit for split-pool barcoding target molecules that are in or on cells or cell organelles
US17/951,008 US11732290B2 (en) 2011-01-31 2022-09-22 Methods of identifying multiple epitopes in cells
US17/951,013 US11667956B2 (en) 2011-01-31 2022-09-22 Methods of identifying multiple epitopes in cells
US18/086,369 US11708599B2 (en) 2011-01-31 2022-12-21 Methods of identifying multiple epitopes in cells
US18/086,383 US11692214B2 (en) 2011-01-31 2022-12-21 Barcoded beads and method for making the same by split-pool synthesis
US18/102,056 US11859240B2 (en) 2011-01-31 2023-01-26 Methods of identifying multiple epitopes in cells
US18/134,003 US11781171B1 (en) 2011-01-31 2023-04-12 Methods of identifying multiple epitopes in cells
US18/233,214 US11932902B2 (en) 2011-01-31 2023-08-11 Barcoded beads and method for making the same by split-pool synthesis
US18/233,221 US11932903B2 (en) 2011-01-31 2023-08-11 Kit for split-pool barcoding target molecules that are in or on cells or cell organelles
US18/379,586 US11926864B1 (en) 2011-01-31 2023-10-12 Method for labeling ligation products with cell-specific barcodes I
US18/379,589 US11939624B2 (en) 2011-01-31 2023-10-12 Method for labeling ligation products with cell-specific barcodes II

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161437854P 2011-01-31 2011-01-31
US61/437,854 2011-01-31
US201161444067P 2011-02-17 2011-02-17
US61/444,067 2011-02-17

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US15/525,876 Continuation-In-Part US20180320241A1 (en) 2014-12-19 2015-12-21 Methods for identifying multiple epitopes in selected sub-populations of cells
PCT/US2015/067147 Continuation-In-Part WO2016100976A2 (en) 2011-01-31 2015-12-21 Methods for identifying multiple epitopes in selected sub-populations of cells

Related Child Applications (4)

Application Number Title Priority Date Filing Date
US13/981,711 A-371-Of-International US10144950B2 (en) 2011-01-31 2012-01-31 Methods of identifying multiple epitopes in cells
US15/597,917 Continuation US10626442B2 (en) 2011-01-31 2017-05-17 Methods of identifying multiple epitopes in cells
US16/147,250 Continuation US10982256B2 (en) 2011-01-31 2018-09-28 Methods of identifying multiple epitopes in cells
US16/163,486 Continuation-In-Part US10982271B2 (en) 2011-01-31 2018-10-17 Methods of identifying multiple epitopes in cells

Publications (2)

Publication Number Publication Date
WO2012106385A2 true WO2012106385A2 (en) 2012-08-09
WO2012106385A3 WO2012106385A3 (en) 2014-03-13

Family

ID=46603278

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/023411 WO2012106385A2 (en) 2011-01-31 2012-01-31 Methods of identifying multiple epitopes in cells

Country Status (6)

Country Link
US (18) US10144950B2 (es)
EP (1) EP2670863B1 (es)
JP (1) JP6069224B2 (es)
CN (1) CN103703143B (es)
ES (1) ES2687761T3 (es)
WO (1) WO2012106385A2 (es)

Cited By (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014026032A2 (en) 2012-08-08 2014-02-13 Apprise Bio, Inc. Increasing dynamic range for identifying multiple epitopes in cells
WO2015047186A1 (en) * 2013-09-30 2015-04-02 Di Wu Methods to profile molecular complexes by using proximity bar-coding
WO2016100976A2 (en) 2014-12-19 2016-06-23 Apprise Bio, Inc. Methods for identifying multiple epitopes in selected sub-populations of cells
WO2016160908A1 (en) * 2015-03-30 2016-10-06 Verily Life Sciences Llc Methods for combining single cell profiling with combinatorial nanoparticle conjugate library screening
WO2017190020A1 (en) * 2016-04-28 2017-11-02 The Scripps Research Institute Oligonucleotide conjugates and uses thereof
WO2018138237A1 (en) 2017-01-27 2018-08-02 Roche Diagnostics Gmbh Barcoded dna for long range sequencing
US20180284125A1 (en) * 2015-03-11 2018-10-04 The Broad Institute, Inc. Proteomic analysis with nucleic acid identifiers
CN108779492A (zh) * 2015-12-30 2018-11-09 生物辐射实验室股份有限公司 数字蛋白质定量
US10144950B2 (en) 2011-01-31 2018-12-04 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US20190187156A1 (en) * 2016-03-22 2019-06-20 California Institute Of Technology Methods for identifying macromolecule interactions
US20190300948A1 (en) * 2016-05-17 2019-10-03 Dname-It Nv Methods for identification of samples
US10501777B2 (en) 2015-07-17 2019-12-10 Nanostring Technologies, Inc. Simultaneous quantification of a plurality of proteins in a user-defined region of a cross-sectioned tissue
US10640816B2 (en) 2015-07-17 2020-05-05 Nanostring Technologies, Inc. Simultaneous quantification of gene expression in a user-defined region of a cross-sectioned tissue
US20210002632A1 (en) * 2011-04-25 2021-01-07 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US11203787B2 (en) 2012-08-13 2021-12-21 The Regents Of The University Of California Methods and systems for detecting biological components
US11312990B2 (en) 2014-06-27 2022-04-26 The Regents Of The University Of California PCR-activated sorting (PAS)
US11332736B2 (en) 2017-12-07 2022-05-17 The Broad Institute, Inc. Methods and compositions for multiplexing single cell and single nuclei sequencing
US11351510B2 (en) 2006-05-11 2022-06-07 Bio-Rad Laboratories, Inc. Microfluidic devices
US11377689B2 (en) 2018-02-12 2022-07-05 Nanostring Technologies, Inc. Chemical compositions and uses thereof
EP3853379A4 (en) * 2018-09-19 2022-07-20 California Institute of Technology HIGH-THROUGH-THROUGH IDENTIFICATION OF INTERMOLECULAR INTERACTIONS
US11408898B2 (en) 2016-12-16 2022-08-09 The Brigham And Women's Hospital, Inc. System, assay and method for partitioning proteins
US11428688B2 (en) 2018-11-07 2022-08-30 Seer, Inc. Compositions, methods and systems for protein corona analysis and uses thereof
US11511242B2 (en) 2008-07-18 2022-11-29 Bio-Rad Laboratories, Inc. Droplet libraries
US11560585B2 (en) * 2011-01-31 2023-01-24 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US11630112B2 (en) 2019-08-05 2023-04-18 Seer, Inc. Systems and methods for sample preparation, data generation, and protein corona analysis
US11634707B2 (en) * 2015-02-10 2023-04-25 Illumina, Inc. Methods and compositions for analyzing cellular components
US11639928B2 (en) 2018-02-22 2023-05-02 10X Genomics, Inc. Methods and systems for characterizing analytes from individual cells or cell populations
US11732287B2 (en) * 2015-02-04 2023-08-22 The Regents Of The University Of California Sequencing of nucleic acids via barcoding in discrete entities
US11732302B2 (en) 2016-12-22 2023-08-22 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11747327B2 (en) 2011-02-18 2023-09-05 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US11819849B2 (en) 2007-02-06 2023-11-21 Brandeis University Manipulation of fluids and reactions in microfluidic systems
US11898193B2 (en) 2011-07-20 2024-02-13 Bio-Rad Laboratories, Inc. Manipulating droplet size
US20240102080A1 (en) * 2014-11-14 2024-03-28 University Of Washington Methods and kits for labeling cellular molecules
US11952626B2 (en) 2021-02-23 2024-04-09 10X Genomics, Inc. Probe-based analysis of nucleic acids and proteins
US11965208B2 (en) * 2019-04-19 2024-04-23 Becton, Dickinson And Company Methods of associating phenotypical data and single cell sequencing data

Families Citing this family (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8592221B2 (en) 2007-04-19 2013-11-26 Brandeis University Manipulation of fluids, fluid components and reactions in microfluidic systems
US8835358B2 (en) 2009-12-15 2014-09-16 Cellular Research, Inc. Digital counting of individual molecules by stochastic attachment of diverse labels
US9399797B2 (en) 2010-02-12 2016-07-26 Raindance Technologies, Inc. Digital analyte analysis
JP5934657B2 (ja) 2010-02-12 2016-06-15 レインダンス テクノロジーズ, インコーポレイテッド デジタル検体分析
US10995362B2 (en) 2011-01-31 2021-05-04 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US10982271B2 (en) 2011-01-31 2021-04-20 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
EP3412778A1 (en) 2011-02-11 2018-12-12 Raindance Technologies, Inc. Methods for forming mixed droplets
DE202012013668U1 (de) 2011-06-02 2019-04-18 Raindance Technologies, Inc. Enzymquantifizierung
ES2663234T3 (es) 2012-02-27 2018-04-11 Cellular Research, Inc Composiciones y kits para recuento molecular
US10752949B2 (en) 2012-08-14 2020-08-25 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10323279B2 (en) 2012-08-14 2019-06-18 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9701998B2 (en) 2012-12-14 2017-07-11 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10400280B2 (en) 2012-08-14 2019-09-03 10X Genomics, Inc. Methods and systems for processing polynucleotides
US11591637B2 (en) 2012-08-14 2023-02-28 10X Genomics, Inc. Compositions and methods for sample processing
US9951386B2 (en) 2014-06-26 2018-04-24 10X Genomics, Inc. Methods and systems for processing polynucleotides
US9567631B2 (en) 2012-12-14 2017-02-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
US10533221B2 (en) 2012-12-14 2020-01-14 10X Genomics, Inc. Methods and systems for processing polynucleotides
CN108753766A (zh) 2013-02-08 2018-11-06 10X基因组学有限公司 多核苷酸条形码生成
DK3611262T3 (da) * 2013-03-15 2021-01-25 Lineage Biosciences Inc Fremgangsmåder til sekvensering af immunrepertoire
EP3842542A1 (en) 2013-08-28 2021-06-30 Becton, Dickinson and Company Massively parallel single cell analysis
US11901041B2 (en) 2013-10-04 2024-02-13 Bio-Rad Laboratories, Inc. Digital analysis of nucleic acid modification
US9944977B2 (en) 2013-12-12 2018-04-17 Raindance Technologies, Inc. Distinguishing rare variations in a nucleic acid sequence from a sample
CN106795553B (zh) 2014-06-26 2021-06-04 10X基因组学有限公司 分析来自单个细胞或细胞群体的核酸的方法
US11098303B2 (en) 2014-07-30 2021-08-24 President And Fellows Of Harvard College Systems and methods for determining nucleic acids
KR102321863B1 (ko) 2015-01-12 2021-11-08 10엑스 제노믹스, 인크. 핵산 시퀀싱 라이브러리의 제조 방법 및 시스템 및 이를 이용하여 제조한 라이브러리
US10697000B2 (en) 2015-02-24 2020-06-30 10X Genomics, Inc. Partition processing methods and systems
US20160298187A1 (en) * 2015-04-08 2016-10-13 Verily Life Sciences Llc Methods of tagging particles for multiplexed functional screening
CA2983937A1 (en) * 2015-04-27 2016-11-03 Abvitro Llc Methods of sequencing, determining, pairing, and validating therapeutic agents and disease specific antigens
US11092607B2 (en) 2015-10-28 2021-08-17 The Board Institute, Inc. Multiplex analysis of single cell constituents
US11371094B2 (en) 2015-11-19 2022-06-28 10X Genomics, Inc. Systems and methods for nucleic acid processing using degenerate nucleotides
US10301677B2 (en) 2016-05-25 2019-05-28 Cellular Research, Inc. Normalization of nucleic acid libraries
SG11201901733PA (en) 2016-09-26 2019-04-29 Cellular Res Inc Measurement of protein expression using reagents with barcoded oligonucleotide sequences
CN116242687A (zh) * 2016-11-08 2023-06-09 哈佛学院院长及董事 使用merfish、扩展显微术和相关技术进行多重成像
CN116908452A (zh) * 2016-12-16 2023-10-20 布里格姆及妇女医院股份有限公司 用于疾病的早期检测的蛋白质冠传感器阵列的系统和方法
US10815525B2 (en) 2016-12-22 2020-10-27 10X Genomics, Inc. Methods and systems for processing polynucleotides
CN117512066A (zh) 2017-01-30 2024-02-06 10X基因组学有限公司 用于基于微滴的单细胞条形编码的方法和系统
ES2968290T3 (es) * 2017-02-02 2024-05-08 New York Genome Center Inc Métodos y composiciones para identificar o cuantificar dianas en una muestra biológica
US10995333B2 (en) 2017-02-06 2021-05-04 10X Genomics, Inc. Systems and methods for nucleic acid preparation
US10844372B2 (en) 2017-05-26 2020-11-24 10X Genomics, Inc. Single cell analysis of transposase accessible chromatin
SG11201901822QA (en) 2017-05-26 2019-03-28 10X Genomics Inc Single cell analysis of transposase accessible chromatin
US11788123B2 (en) 2017-05-26 2023-10-17 President And Fellows Of Harvard College Systems and methods for high-throughput image-based screening
CN111386351A (zh) 2017-09-22 2020-07-07 华盛顿大学 细胞分子的原位组合标记
SG11201913654QA (en) 2017-11-15 2020-01-30 10X Genomics Inc Functionalized gel beads
US10829815B2 (en) 2017-11-17 2020-11-10 10X Genomics, Inc. Methods and systems for associating physical and genetic properties of biological particles
EP3752832A1 (en) 2018-02-12 2020-12-23 10X Genomics, Inc. Methods characterizing multiple analytes from individual cells or cell populations
CN112262218A (zh) 2018-04-06 2021-01-22 10X基因组学有限公司 用于单细胞处理中的质量控制的系统和方法
US20200032335A1 (en) 2018-07-27 2020-01-30 10X Genomics, Inc. Systems and methods for metabolome analysis
CN112805389A (zh) 2018-10-01 2021-05-14 贝克顿迪金森公司 确定5’转录物序列
US11459607B1 (en) 2018-12-10 2022-10-04 10X Genomics, Inc. Systems and methods for processing-nucleic acid molecules from a single cell using sequential co-partitioning and composite barcodes
EP3894593A2 (en) 2018-12-13 2021-10-20 DNA Script Direct oligonucleotide synthesis on cells and biomolecules
WO2020139757A1 (en) * 2018-12-27 2020-07-02 Bio-Rad Laboratories, Inc. Sequential multiplex western blotting
US11845983B1 (en) 2019-01-09 2023-12-19 10X Genomics, Inc. Methods and systems for multiplexing of droplet based assays
EP3914728B1 (en) 2019-01-23 2023-04-05 Becton, Dickinson and Company Oligonucleotides associated with antibodies
WO2020185312A2 (en) 2019-01-25 2020-09-17 President And Fellows Of Harvard College Compositions and method for synthesizing nucleic acids
US11634766B2 (en) * 2019-02-04 2023-04-25 The Broad Institute, Inc. Methods and compositions for analyzing nucleic acids
US11851683B1 (en) 2019-02-12 2023-12-26 10X Genomics, Inc. Methods and systems for selective analysis of cellular samples
EP4004231A1 (en) 2019-07-22 2022-06-01 Becton, Dickinson and Company Single cell chromatin immunoprecipitation sequencing assay
CN114729350A (zh) 2019-11-08 2022-07-08 贝克顿迪金森公司 使用随机引发获得用于免疫组库测序的全长v(d)j信息
CN110819656B (zh) * 2019-11-11 2021-06-04 中国科学院上海高等研究院 一种基于同步光源的x-射线多色遗传标记探针及其制备方法以及应用
WO2021146207A1 (en) 2020-01-13 2021-07-22 Becton, Dickinson And Company Methods and compositions for quantitation of proteins and rna
EP4090464A1 (en) 2020-01-17 2022-11-23 F. Hoffmann-La Roche AG Microfluidic device and method for automated split-pool synthesis
EP4121528A1 (en) 2020-03-17 2023-01-25 F. Hoffmann-La Roche AG Method of improving cell recovery in single-cell analysis
US20230166259A1 (en) 2020-05-01 2023-06-01 Roche Sequencing Solutions, Inc. Sample collection tray for multi-well plates
US20230242971A1 (en) 2020-05-08 2023-08-03 Roche Sequencing Solutions, Inc. Removal of excess oligonucleotides from a reation mixture
CN115605614A (zh) 2020-05-14 2023-01-13 贝克顿迪金森公司(Us) 用于免疫组库谱分析的引物
WO2021249816A1 (en) 2020-06-08 2021-12-16 F. Hoffmann-La Roche Ag Method of single-cell analysis of multiple samples
WO2022008649A1 (en) 2020-07-08 2022-01-13 F. Hoffmann-La Roche Ag Spatial analysis of multiple targets in tissue samples
US20230249178A1 (en) 2020-07-08 2023-08-10 Roche Sequencing Solutions, Inc. Split-pool synthesis apparatus and methods of performing split-pool synthesis
US11932901B2 (en) 2020-07-13 2024-03-19 Becton, Dickinson And Company Target enrichment using nucleic acid probes for scRNAseq
EP4247967A1 (en) 2020-11-20 2023-09-27 Becton, Dickinson and Company Profiling of highly expressed and lowly expressed proteins
US20220177950A1 (en) 2020-12-03 2022-06-09 Roche Sequencing Solutions, Inc. Whole transcriptome analysis in single cells
WO2022164893A1 (en) * 2021-01-26 2022-08-04 Chan Zuckerberg Biohub, Inc. Quantification of cellular proteins using barcoded binding moieties
CN113189078B (zh) * 2021-03-04 2024-04-16 吉林大学 一种靶向药物的高通量筛选方法
WO2023086847A1 (en) 2021-11-10 2023-05-19 Encodia, Inc. Methods for barcoding macromolecules in individual cells
EP4206674A1 (en) 2021-12-28 2023-07-05 Encodia, Inc. High-throughput serotyping and antibody profiling assays

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5831012A (en) 1994-01-14 1998-11-03 Pharmacia & Upjohn Aktiebolag Bacterial receptor structures
US6262490B1 (en) 1999-11-05 2001-07-17 Advanced Semiconductor Engineering, Inc. Substrate strip for use in packaging semiconductor chips
US20030219801A1 (en) 2002-03-06 2003-11-27 Affymetrix, Inc. Aptamer base technique for ligand identification
US6670461B1 (en) 1997-09-12 2003-12-30 Exiqon A/S Oligonucleotide analogues
US6770748B2 (en) 1997-03-07 2004-08-03 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogue
US6830936B2 (en) 1995-06-29 2004-12-14 Affymetrix Inc. Integrated nucleic acid diagnostic device
US20070166708A1 (en) 2001-07-03 2007-07-19 Krassen Dimitrov Methods for detection and quantification of analytes in complex mixtures
US7294503B2 (en) 2000-09-15 2007-11-13 California Institute Of Technology Microfabricated crossflow devices and methods
US7294298B2 (en) 2002-07-24 2007-11-13 Tdk Corporation Functional film for transfer having functional layer, object furnished with functional layer and process for producing the same
US7378280B2 (en) 2000-11-16 2008-05-27 California Institute Of Technology Apparatus and methods for conducting assays and high throughput screening

Family Cites Families (115)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4568649A (en) 1983-02-22 1986-02-04 Immunex Corporation Immediate ligand detection assay
US5242794A (en) 1984-12-13 1993-09-07 Applied Biosystems, Inc. Detection of specific sequences in nucleic acids
CA1323293C (en) 1987-12-11 1993-10-19 Keith C. Backman Assay using template-dependent nucleic acid probe reorganization
US4988617A (en) 1988-03-25 1991-01-29 California Institute Of Technology Method of detecting a nucleotide change in nucleic acids
EP0425563B1 (en) 1988-07-20 1996-05-15 David Segev Process for amplifying and detecting nucleic acid sequences
US5185243A (en) 1988-08-25 1993-02-09 Syntex (U.S.A.) Inc. Method for detection of specific nucleic acid sequences
DE69118930T2 (de) 1990-01-26 1997-01-09 Abbott Lab Verbessertes Verfahren zur Amplifikation von Nuklein säurezielsequenz, einsetzbar für die Polymerase und Ligasekettenreaktion
US5494810A (en) 1990-05-03 1996-02-27 Cornell Research Foundation, Inc. Thermostable ligase-mediated DNA amplifications system for the detection of genetic disease
DE69620596T2 (de) 1995-05-19 2002-11-21 Abbott Lab Nachweis von nukleinsäuren mit breitem dynamikbereich unter verwendung von aggregat-primer-reihen
US6969488B2 (en) 1998-05-22 2005-11-29 Solexa, Inc. System and apparatus for sequential processing of analytes
US6974669B2 (en) * 2000-03-28 2005-12-13 Nanosphere, Inc. Bio-barcodes based on oligonucleotide-modified nanoparticles
US6607878B2 (en) * 1997-10-06 2003-08-19 Stratagene Collections of uniquely tagged molecules
US6180340B1 (en) 1997-10-31 2001-01-30 Gen-Probe Incorporated Extended dynamic range assays
US6787308B2 (en) 1998-07-30 2004-09-07 Solexa Ltd. Arrayed biomolecules and their use in sequencing
US20030022207A1 (en) 1998-10-16 2003-01-30 Solexa, Ltd. Arrayed polynucleotides and their use in genome analysis
US20040106110A1 (en) 1998-07-30 2004-06-03 Solexa, Ltd. Preparation of polynucleotide arrays
US6221613B1 (en) 1998-12-31 2001-04-24 Synaptic Pharmaceutical Corporation DNA encoding a human melanin concentrating hormone receptor (MCH1) and uses thereof
US7056661B2 (en) 1999-05-19 2006-06-06 Cornell Research Foundation, Inc. Method for sequencing nucleic acid molecules
ATE320443T1 (de) 1999-07-15 2006-04-15 Qiagen Gmbh Verfahren zur trennung von teilchenförmigen substraten aus einer lösung indem man den teilchenverlust minimiert
US7211390B2 (en) 1999-09-16 2007-05-01 454 Life Sciences Corporation Method of sequencing a nucleic acid
US7244559B2 (en) 1999-09-16 2007-07-17 454 Life Sciences Corporation Method of sequencing a nucleic acid
EP1218543A2 (en) 1999-09-29 2002-07-03 Solexa Ltd. Polynucleotide sequencing
US6582938B1 (en) 2001-05-11 2003-06-24 Affymetrix, Inc. Amplification of nucleic acids
GB0002389D0 (en) 2000-02-02 2000-03-22 Solexa Ltd Molecular arrays
AU2001234779A1 (en) 2000-02-03 2001-08-14 Nanoscale Combinatorial Synthesis, Inc. Structure identification methods using mass measurements
CA2401962A1 (en) 2000-02-07 2001-08-09 Illumina, Inc. Nucleic acid detection methods using universal priming
SE516272C2 (sv) 2000-02-18 2001-12-10 Ulf Landegren Metoder och kit för analytdetektion mha proximitets-probning
WO2002027029A2 (en) 2000-09-27 2002-04-04 Lynx Therapeutics, Inc. Method for determining relative abundance of nucleic acid sequences
DE10120797B4 (de) 2001-04-27 2005-12-22 Genovoxx Gmbh Verfahren zur Analyse von Nukleinsäureketten
DE10239504A1 (de) 2001-08-29 2003-04-24 Genovoxx Gmbh Verfahren zur Analyse von Nukleinsäurekettensequenzen und der Genexpression
JP2003101204A (ja) 2001-09-25 2003-04-04 Nec Kansai Ltd 配線基板及び配線基板の製造方法並びに電子部品
US20030082551A1 (en) 2001-09-28 2003-05-01 Zarling David A. High-throughput gene cloning and phenotypic screening
US20050227231A1 (en) 2001-10-04 2005-10-13 Dimitri Tcherkassov Device for sequencing nucleic acid molecules
DE10149786B4 (de) 2001-10-09 2013-04-25 Dmitry Cherkasov Oberfläche für Untersuchungen aus Populationen von Einzelmolekülen
US20050124022A1 (en) 2001-10-30 2005-06-09 Maithreyan Srinivasan Novel sulfurylase-luciferase fusion proteins and thermostable sulfurylase
US6902921B2 (en) 2001-10-30 2005-06-07 454 Corporation Sulfurylase-luciferase fusion proteins and thermostable sulfurylase
DE10214395A1 (de) 2002-03-30 2003-10-23 Dmitri Tcherkassov Verfahren zur Analyse von Einzelnukleotidpolymorphismen
US7108976B2 (en) 2002-06-17 2006-09-19 Affymetrix, Inc. Complexity management of genomic DNA by locus specific amplification
GB0222846D0 (en) * 2002-10-03 2002-11-06 Choo Yen Cell culture
DE10356937A1 (de) 2002-12-09 2004-07-15 Mold-Masters Ltd., Georgetown Düsenspitze und -dichtung
US20040110153A1 (en) 2002-12-10 2004-06-10 Affymetrix, Inc. Compleixity management of genomic DNA by semi-specific amplification
WO2004070007A2 (en) 2003-01-29 2004-08-19 454 Corporation Method for preparing single-stranded dna libraries
ATE418621T1 (de) 2003-04-24 2009-01-15 Afshin Ahmadian Verfahren zur detektion allelspezifischer mutationen
WO2005010159A2 (en) * 2003-07-17 2005-02-03 Children's Hospital Medical Center Rolling circle amplification of micro-rna samples
DE10356837A1 (de) 2003-12-05 2005-06-30 Dmitry Cherkasov Modifizierte Nukleotide und Nukleoside
EP1725572B1 (de) 2003-11-05 2017-05-31 AGCT GmbH Makromolekulare nukleotidverbindungen und methoden zu deren anwendung
US7169560B2 (en) 2003-11-12 2007-01-30 Helicos Biosciences Corporation Short cycle methods for sequencing polynucleotides
WO2005050224A2 (en) 2003-11-13 2005-06-02 Epitome Biosystems Inc. Small molecule and peptide arrays and uses thereof
US20080261204A1 (en) 2004-01-23 2008-10-23 Lingvitae As Polynucleotide Ligation Reactions
DE102004009704A1 (de) 2004-02-27 2005-09-15 Dmitry Cherkasov Makromolekulare Nukleotidverbindungen und Methoden zu deren Anwendung
US20060275923A1 (en) 2004-03-23 2006-12-07 Hammond David J Methods for reducing the range in concentrations of analyte species in a sample
DE102004025696A1 (de) 2004-05-26 2006-02-23 Dmitry Cherkasov Verfahren, Oberfläche und Substrate zu hochparallelen Analysen von Nukleinsäureketten
DE102004025694A1 (de) 2004-05-26 2006-02-23 Dmitry Cherkasov Verfahren und Oberfläche zu hochparallelen Analysen von Nukleinsäureketten
DE102004025744A1 (de) 2004-05-26 2005-12-29 Dmitry Cherkasov Oberfläche für die Analysen an einzelnen Nukleinsäuremolekülen
DE102004025695A1 (de) 2004-05-26 2006-02-23 Dmitry Cherkasov Verfahren und Oberfläche zur parallelen Sequenzierung von Nukleinsäureketten
DE102004025745A1 (de) 2004-05-26 2005-12-15 Cherkasov, Dmitry Oberfläche für die Analysen an einzelnen Molekülen
DE102004025746A1 (de) 2004-05-26 2005-12-15 Dmitry Cherkasov Verfahren, Oberfläche und Substrate zur hochparallelen Sequenzierung von Nukleinsäureketten
WO2005123963A2 (en) 2004-06-14 2005-12-29 The Board Of Trustees Of The Leland Stanford Junior University Methods and compositions for use in analyte detection using proximity probes
US20060002471A1 (en) 2004-06-30 2006-01-05 Lippincott Louis A Motion estimation unit
US20060024711A1 (en) 2004-07-02 2006-02-02 Helicos Biosciences Corporation Methods for nucleic acid amplification and sequence determination
US7276720B2 (en) 2004-07-19 2007-10-02 Helicos Biosciences Corporation Apparatus and methods for analyzing samples
US20060012793A1 (en) 2004-07-19 2006-01-19 Helicos Biosciences Corporation Apparatus and methods for analyzing samples
US20060024678A1 (en) 2004-07-28 2006-02-02 Helicos Biosciences Corporation Use of single-stranded nucleic acid binding proteins in sequencing
WO2006097320A2 (de) 2005-03-17 2006-09-21 Genovoxx Gmbh Makromolekulare nukleotidverbindungen und methoden zu deren anwendung
US20060246475A1 (en) 2005-01-21 2006-11-02 Third Wave Technologies Compositions and methods for increased dynamic range detection of nucleic acids
DE102006012317A1 (de) 2005-03-17 2007-01-18 Cherkasov, Dmitry, Dr. Makromolekulare Nukleotidverbindungen und Methoden zu deren Anwendung
WO2006138257A2 (en) 2005-06-15 2006-12-28 Callida Genomics, Inc. Single molecule arrays for genetic and chemical analysis
AU2007249635B2 (en) 2005-10-07 2012-05-31 Complete Genomics, Inc. High throughput genome sequencing on DNA arrays
US8986926B2 (en) 2005-12-23 2015-03-24 Nanostring Technologies, Inc. Compositions comprising oriented, immobilized macromolecules and methods for their preparation
CA2640385C (en) 2005-12-23 2014-07-15 Nanostring Technologies, Inc. Nanoreporters and methods of manufacturing and use thereof
US8394590B2 (en) * 2006-08-02 2013-03-12 California Institute Of Technology Capture agents and related methods and systems for detecting and/or sorting targets
WO2008061193A2 (en) 2006-11-15 2008-05-22 Biospherex Llc Multitag sequencing and ecogenomics analysis
US20080269068A1 (en) 2007-02-06 2008-10-30 President And Fellows Of Harvard College Multiplex decoding of sequence tags in barcodes
US8003312B2 (en) * 2007-02-16 2011-08-23 The Board Of Trustees Of The Leland Stanford Junior University Multiplex cellular assays using detectable cell barcodes
US8090603B2 (en) 2007-05-11 2012-01-03 Fansnap, Inc. System and method for selecting event tickets
WO2008155599A1 (en) 2007-06-20 2008-12-24 Taag-Genetics Sa Nested multiplex amplification method for identification of multiple biological entities
US8999652B2 (en) 2007-08-10 2015-04-07 Primity Bio, Inc. Biological encoding of large numbers of cells
EP2203749B1 (en) 2007-10-05 2012-08-29 Affymetrix, Inc. Highly multiplexed particle-based assays
EP2247708A4 (en) 2007-12-17 2013-02-27 Yeda Res & Dev SYSTEM AND METHOD FOR PROCESSING AND MANIPULATING DNA
EP2279291B1 (en) 2008-04-09 2019-07-03 Cornell University Coferons and methods of making and using them
DE102008025992B4 (de) 2008-05-30 2011-01-27 Siemens Healthcare Diagnostics Gmbh Titerplatte und Verfahren zur Detektion eines Analyten
JP5836803B2 (ja) 2008-08-14 2015-12-24 ナノストリング テクノロジーズ, インコーポレイテッド 安定したナノレポーター
WO2010027870A2 (en) * 2008-08-26 2010-03-11 Fluidigm Corporation Assay methods for increased throughput of samples and/or targets
DE102008044946B4 (de) 2008-08-29 2022-06-15 Evonik Superabsorber Gmbh Einsatz von Schaumkörpern in Oxidations-Reaktoren zur Herstellung ungesättigter Carbonsäuren
GB0816976D0 (en) 2008-09-16 2008-10-22 Natural Environment Res Council Protein
US8748103B2 (en) 2008-11-07 2014-06-10 Sequenta, Inc. Monitoring health and disease status using clonotype profiles
US8481698B2 (en) 2009-03-19 2013-07-09 The President And Fellows Of Harvard College Parallel proximity ligation event analysis
CN102203281B (zh) 2009-08-21 2013-04-17 赛业(广州)生物科技有限公司 复制条码筛选检测
US8835358B2 (en) 2009-12-15 2014-09-16 Cellular Research, Inc. Digital counting of individual molecules by stochastic attachment of diverse labels
US8648016B2 (en) 2010-02-08 2014-02-11 Robert Bosch Gmbh Array with extended dynamic range and associated method
US20140186827A1 (en) 2010-05-14 2014-07-03 Fluidigm, Inc. Assays for the detection of genotype, mutations, and/or aneuploidy
WO2011146942A1 (en) 2010-05-21 2011-11-24 The Translational Genomics Research Institute Methods and kits to analyze microrna by nucleic acid sequencing
US9696332B2 (en) 2010-07-23 2017-07-04 Matrix Technologies Llc Automated liquid handling device
EP2622103B2 (en) * 2010-09-30 2022-11-16 Bio-Rad Laboratories, Inc. Sandwich assays in droplets
EP3561159B1 (en) * 2010-10-08 2023-09-20 President and Fellows of Harvard College High-throughput single cell barcoding
LT3360963T (lt) 2010-11-12 2020-02-25 Gen9, Inc. Nukleorūgščių sintezės būdai ir įrenginiai
EP2652155B1 (en) 2010-12-16 2016-11-16 Gigagen, Inc. Methods for massively parallel analysis of nucleic acids in single cells
US11560585B2 (en) 2011-01-31 2023-01-24 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US10982271B2 (en) 2011-01-31 2021-04-20 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US20200157603A1 (en) 2011-01-31 2020-05-21 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US10995362B2 (en) 2011-01-31 2021-05-04 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
JP6069224B2 (ja) 2011-01-31 2017-02-01 アプライズ バイオ, インコーポレイテッド 細胞において複数のエピトープを同定する方法
AU2011358564B9 (en) 2011-02-09 2017-07-13 Natera, Inc Methods for non-invasive prenatal ploidy calling
WO2012142242A2 (en) 2011-04-12 2012-10-18 Siloam Biosciences, Inc. Assay method for extended dynamic range
WO2013137737A1 (en) 2012-03-16 2013-09-19 Flexgen B.V. Methods of creating and screening of dna encoded combinatorial libraries of chemical antibodies
JP6525872B2 (ja) 2012-08-08 2019-06-05 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft 細胞中の複数のエピトープを同定するためのダイナミックレンジを高めること
CN103730314A (zh) 2012-10-12 2014-04-16 无锡慧明电子科技有限公司 一种气体离子源进气实时控制系统
MX2015007743A (es) 2012-12-17 2015-12-07 Harvard College Ingenieria del genoma humano guiada por ácido robonucleico.
WO2015035087A1 (en) 2013-09-04 2015-03-12 Fluidigm Corporation Proximity assays for detecting nucleic acids and proteins in a single cell
GB201401885D0 (en) 2014-02-04 2014-03-19 Olink Ab Proximity assay with detection based on hybridisation chain reaction (HCR)
US10900065B2 (en) 2014-11-14 2021-01-26 University Of Washington Methods and kits for labeling cellular molecules
JP2017537645A (ja) 2014-12-19 2017-12-21 アプライズ バイオ, インコーポレイテッド 細胞の選択された部分集団中の複数エピトープを識別するための方法
CN206096158U (zh) 2016-09-19 2017-04-12 清华大学 自动液体处理系统
US20180089373A1 (en) 2016-09-23 2018-03-29 Driver, Inc. Integrated systems and methods for automated processing and analysis of biological samples, clinical information processing and clinical trial matching
US20180333851A1 (en) 2017-05-19 2018-11-22 Counsyl, Inc. Modular robotic system for laboratory diagnostics

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5831012A (en) 1994-01-14 1998-11-03 Pharmacia & Upjohn Aktiebolag Bacterial receptor structures
US6830936B2 (en) 1995-06-29 2004-12-14 Affymetrix Inc. Integrated nucleic acid diagnostic device
US6770748B2 (en) 1997-03-07 2004-08-03 Takeshi Imanishi Bicyclonucleoside and oligonucleotide analogue
US6670461B1 (en) 1997-09-12 2003-12-30 Exiqon A/S Oligonucleotide analogues
US6794499B2 (en) 1997-09-12 2004-09-21 Exiqon A/S Oligonucleotide analogues
US6262490B1 (en) 1999-11-05 2001-07-17 Advanced Semiconductor Engineering, Inc. Substrate strip for use in packaging semiconductor chips
US7294503B2 (en) 2000-09-15 2007-11-13 California Institute Of Technology Microfabricated crossflow devices and methods
US7378280B2 (en) 2000-11-16 2008-05-27 California Institute Of Technology Apparatus and methods for conducting assays and high throughput screening
US20070166708A1 (en) 2001-07-03 2007-07-19 Krassen Dimitrov Methods for detection and quantification of analytes in complex mixtures
US7473767B2 (en) 2001-07-03 2009-01-06 The Institute For Systems Biology Methods for detection and quantification of analytes in complex mixtures
US20030219801A1 (en) 2002-03-06 2003-11-27 Affymetrix, Inc. Aptamer base technique for ligand identification
US7294298B2 (en) 2002-07-24 2007-11-13 Tdk Corporation Functional film for transfer having functional layer, object furnished with functional layer and process for producing the same

Non-Patent Citations (33)

* Cited by examiner, † Cited by third party
Title
"The Electronic Notebook", 2000, CRC PRESS, article "Basic Methods in Antibody Production and Characterization"
A. JOHNSTONE; M. TURNER: "Immunochemistry", vol. 1-2, 1997, OXFORD UNIVERSITY PRESS
A. JOHNSTONE; R. THORPE: "Immunochemistry in Practice", 1996, BLACKWELL SCIENCE, LTD.
B. WLOTZKA ET AL., PROC. NATL. ACAD. SCI., vol. 99, 2002, pages 8898 - 8902
BOOZER ET AL., ANALYTICAL CHEMISTRY, vol. 76, no. 23, 2004, pages 6967 - 6972
C. BORREBAECK: "Antibody Engineering", 1995, OXFORD UNIVERSITY PRESS
COLIGAN ET AL: "Current Protocols in Immunology", 1999, JOHN WILEY & SONS
D. WILSON; J. SZOSTAK, ANN. REV. BIOCHEM., vol. 68, 1999, pages 611 - 647
E. HARLOW; D. LANE: "Using Antibodies", 1999, COLD SPRING HARBOR LAB PRESS
GOLDMAN ET AL., J. AM. CHEM. SOC., vol. 124, 2002, pages 6378 - 6382
GULLBERG ET AL., PNAS, vol. 101, no. 22, 2004, pages 228420 - 228424
H. ZOLA: "Monoclonal Antibodies: Preparation and Use of Monoclonal Antibodies and Engineered Antibody Derivatives (Basics: From Background to Bench", 2000, SPRINGER VERLAG
J. GODING: "Monoclonal Antibodies: Principles and Practice", 1996, ACADEMIC PRESS
J. MAYNARD; G. GEORGIOU, ANN. REV. BIOMED. ENG., vol. 2, no. 339, 2000, pages 76
KOZLOV ET AL., BIOPOLYMERS 5, vol. 73, no. 5, 2004, pages 621 - 630
L. GOLD, J. BIOL. CHEM., vol. 270, no. 23, 1995, pages 13581 - 13584
L. JERMUTUS ET AL., EUR. BIOPHYS. J., vol. 31, 2002, pages 179 - 184
MANIATIS, T.: "Molecular Cloning", 1982, COLD SPRING HARBOR LABORATORY
NATURE BIOTECHNOLOGY, vol. 20, pages 473 - 477
NOLAN, NATURE METHODS, vol. 2, 2005, pages 11 - 12
P. SHEPHERD; C. DEAN: "Monoclonal Antibodies: A Practical Approach", 2000, OXFORD UNIVERSITY PRESS
PATHAK ET AL., J. AM. CHEM. SOC., vol. 123, 2001, pages 4103 - 4104
R. KONTERMANN AND S. DUBEL: "Springer Lab Manual", 2001, SPRINGER VERLAG, article "Antibody Engineering"
REMADE ET AL., PROC. NATL. SCI. USA, vol. 18, 2000, pages 553 - 558
S S. SPADA ET AL., BIOL. CHEM., vol. 378, 1997, pages 445 - 456
S. HOCKFIELD ET AL.: "Selected Methods for Antibody and Nucleic Acid Probes", 1993, COLD SPRING HARBOR LAB PRESS
S. JAYASENA, CLIN. CHEM., vol. 45, 1999, pages 1628 - 1650
S. PAUL: "Antibody Engineering Protocols", 1995, HUMANA PRESS
SCIENCE, vol. 301, 2003, pages 1884 - 1886
SIMON ET AL., PNAS, vol. 15; 89, no. 20, 1992, pages 9367 - 9371
TANNER ET AL., SPECTROCHIMICA ACTA PART B: ATOMIC SPECTROSCOPY, vol. 62, no. 3, 2007, pages 188 - 195
TURRO, N. J.: "Modern Molecular Photochemistry", 1978, BENJAMIN/CUMMINGS PUBLISHING COL, INC., pages: 296 - 361
V. SIEBER ET AL., NAT BIOTECHNOL., vol. 16, no. 10, 1998, pages 955 - 960

Cited By (78)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11351510B2 (en) 2006-05-11 2022-06-07 Bio-Rad Laboratories, Inc. Microfluidic devices
US11819849B2 (en) 2007-02-06 2023-11-21 Brandeis University Manipulation of fluids and reactions in microfluidic systems
US11596908B2 (en) 2008-07-18 2023-03-07 Bio-Rad Laboratories, Inc. Droplet libraries
US11511242B2 (en) 2008-07-18 2022-11-29 Bio-Rad Laboratories, Inc. Droplet libraries
US11534727B2 (en) 2008-07-18 2022-12-27 Bio-Rad Laboratories, Inc. Droplet libraries
US11692214B2 (en) 2011-01-31 2023-07-04 Roche Sequencing Solutions, Inc. Barcoded beads and method for making the same by split-pool synthesis
US11781171B1 (en) 2011-01-31 2023-10-10 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US11667956B2 (en) 2011-01-31 2023-06-06 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US11634752B2 (en) 2011-01-31 2023-04-25 Roche Sequencing Solutions, Inc. Kit for split-pool barcoding target molecules that are in or on cells or cell organelles
US11859240B2 (en) 2011-01-31 2024-01-02 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US11708599B2 (en) 2011-01-31 2023-07-25 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US11566278B2 (en) 2011-01-31 2023-01-31 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US11939624B2 (en) 2011-01-31 2024-03-26 Roche Sequencing Solutions, Inc. Method for labeling ligation products with cell-specific barcodes II
US11560585B2 (en) * 2011-01-31 2023-01-24 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US11732290B2 (en) 2011-01-31 2023-08-22 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US10144950B2 (en) 2011-01-31 2018-12-04 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US10626442B2 (en) 2011-01-31 2020-04-21 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US11512341B1 (en) 2011-01-31 2022-11-29 Roche Sequencing Solutions, Inc. Methods of identifying multiple epitopes in cells
US11932903B2 (en) 2011-01-31 2024-03-19 Roche Sequencing Solutions, Inc. Kit for split-pool barcoding target molecules that are in or on cells or cell organelles
US11932902B2 (en) 2011-01-31 2024-03-19 Roche Sequencing Solutions, Inc. Barcoded beads and method for making the same by split-pool synthesis
US11926864B1 (en) 2011-01-31 2024-03-12 Roche Sequencing Solutions, Inc. Method for labeling ligation products with cell-specific barcodes I
US11747327B2 (en) 2011-02-18 2023-09-05 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US11965877B2 (en) 2011-02-18 2024-04-23 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US11768198B2 (en) 2011-02-18 2023-09-26 Bio-Rad Laboratories, Inc. Compositions and methods for molecular labeling
US20210002632A1 (en) * 2011-04-25 2021-01-07 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US11939573B2 (en) * 2011-04-25 2024-03-26 Bio-Rad Laboratories, Inc. Methods and compositions for nucleic acid analysis
US11898193B2 (en) 2011-07-20 2024-02-13 Bio-Rad Laboratories, Inc. Manipulating droplet size
CN105264088B (zh) * 2012-08-08 2018-12-28 豪夫迈·罗氏有限公司 提高鉴定细胞中的多个表位的动态范围
EP3578669A1 (en) * 2012-08-08 2019-12-11 F. Hoffmann-La Roche AG Increasing dynamic range for identifying multiple epitopes in cells
WO2014026032A3 (en) * 2012-08-08 2015-04-30 Apprise Bio, Inc. Increasing dynamic range for identifying multiple epitopes in cells
JP2015525575A (ja) * 2012-08-08 2015-09-07 アプライズ バイオ, インコーポレイテッド 細胞中の複数のエピトープを同定するためのダイナミックレンジを高めること
CN105264088A (zh) * 2012-08-08 2016-01-20 爱普瑞斯生物公司 提高鉴定细胞中的多个表位的动态范围
US10174310B2 (en) 2012-08-08 2019-01-08 Roche Sequencing Solutions, Inc. Increasing dynamic range for identifying multiple epitopes in cells
WO2014026032A2 (en) 2012-08-08 2014-02-13 Apprise Bio, Inc. Increasing dynamic range for identifying multiple epitopes in cells
US11891666B2 (en) 2012-08-13 2024-02-06 The Regents Of The University Of California Methods and systems for detecting biological components
US11203787B2 (en) 2012-08-13 2021-12-21 The Regents Of The University Of California Methods and systems for detecting biological components
EP3052658A4 (en) * 2013-09-30 2017-03-15 Di Wu Methods to profile molecular complexes by using proximity dependant bar-coding
WO2015047186A1 (en) * 2013-09-30 2015-04-02 Di Wu Methods to profile molecular complexes by using proximity bar-coding
US10982257B2 (en) 2013-09-30 2021-04-20 Vesicode Ab Methods to profile molecular complexes or single cells via proximity dependent barcoding
US20200248235A1 (en) * 2013-09-30 2020-08-06 Di Wu Methods to profile molecular complexes or single cells via by using proximity bar-coding dependant barcoding
US10633691B2 (en) 2013-09-30 2020-04-28 Di Wu Methods to profile molecular complexes or single cells via proximity dependant barcoding
CN105745334A (zh) * 2013-09-30 2016-07-06 吴迪 通过使用邻近条形编码分析分子复合物的概况的方法
US20160281134A1 (en) * 2013-09-30 2016-09-29 Di Wu Methods to profile molecular complexes or single cells via proximity dependant barcoding
US11312990B2 (en) 2014-06-27 2022-04-26 The Regents Of The University Of California PCR-activated sorting (PAS)
US20240102080A1 (en) * 2014-11-14 2024-03-28 University Of Washington Methods and kits for labeling cellular molecules
WO2016100976A2 (en) 2014-12-19 2016-06-23 Apprise Bio, Inc. Methods for identifying multiple epitopes in selected sub-populations of cells
EP3234602A4 (en) * 2014-12-19 2018-05-30 H. Hoffnabb-La Roche Ag Methods for identifying multiple epitopes in selected sub-populations of cells
US11732287B2 (en) * 2015-02-04 2023-08-22 The Regents Of The University Of California Sequencing of nucleic acids via barcoding in discrete entities
US11634707B2 (en) * 2015-02-10 2023-04-25 Illumina, Inc. Methods and compositions for analyzing cellular components
US20180284125A1 (en) * 2015-03-11 2018-10-04 The Broad Institute, Inc. Proteomic analysis with nucleic acid identifiers
WO2016160908A1 (en) * 2015-03-30 2016-10-06 Verily Life Sciences Llc Methods for combining single cell profiling with combinatorial nanoparticle conjugate library screening
US11708602B2 (en) 2015-07-17 2023-07-25 Nanostring Technologies, Inc. Simultaneous quantification of gene expression in a user-defined region of a cross-sectioned tissue
US10501777B2 (en) 2015-07-17 2019-12-10 Nanostring Technologies, Inc. Simultaneous quantification of a plurality of proteins in a user-defined region of a cross-sectioned tissue
US10640816B2 (en) 2015-07-17 2020-05-05 Nanostring Technologies, Inc. Simultaneous quantification of gene expression in a user-defined region of a cross-sectioned tissue
US20230417765A1 (en) * 2015-12-30 2023-12-28 Bio-Rad Laboratories, Inc. Digital protein quantification
CN108779492A (zh) * 2015-12-30 2018-11-09 生物辐射实验室股份有限公司 数字蛋白质定量
US11965891B2 (en) * 2015-12-30 2024-04-23 Bio-Rad Laboratories, Inc. Digital protein quantification
US10900974B2 (en) * 2016-03-22 2021-01-26 California Institute Of Technology Methods for identifying macromolecule interactions
US20190187156A1 (en) * 2016-03-22 2019-06-20 California Institute Of Technology Methods for identifying macromolecule interactions
WO2017190020A1 (en) * 2016-04-28 2017-11-02 The Scripps Research Institute Oligonucleotide conjugates and uses thereof
US20190300948A1 (en) * 2016-05-17 2019-10-03 Dname-It Nv Methods for identification of samples
US11435360B2 (en) 2016-12-16 2022-09-06 The Brigham And Women's Hospital, Inc. System and sensor array
US11567086B2 (en) 2016-12-16 2023-01-31 The Brigham And Women's Hospital, Inc. System and method for protein corona sensor array for early detection of diseases
US11408898B2 (en) 2016-12-16 2022-08-09 The Brigham And Women's Hospital, Inc. System, assay and method for partitioning proteins
US11732302B2 (en) 2016-12-22 2023-08-22 10X Genomics, Inc. Methods and systems for processing polynucleotides
WO2018138237A1 (en) 2017-01-27 2018-08-02 Roche Diagnostics Gmbh Barcoded dna for long range sequencing
CN110291207A (zh) * 2017-01-27 2019-09-27 豪夫迈·罗氏有限公司 用于长程测序的条形码化dna
US11332736B2 (en) 2017-12-07 2022-05-17 The Broad Institute, Inc. Methods and compositions for multiplexing single cell and single nuclei sequencing
US11473142B2 (en) 2018-02-12 2022-10-18 Nanostring Technologies, Inc. Chemical compositions and uses thereof
US11377689B2 (en) 2018-02-12 2022-07-05 Nanostring Technologies, Inc. Chemical compositions and uses thereof
US11639928B2 (en) 2018-02-22 2023-05-02 10X Genomics, Inc. Methods and systems for characterizing analytes from individual cells or cell populations
US11852628B2 (en) 2018-02-22 2023-12-26 10X Genomics, Inc. Methods and systems for characterizing analytes from individual cells or cell populations
EP3853379A4 (en) * 2018-09-19 2022-07-20 California Institute of Technology HIGH-THROUGH-THROUGH IDENTIFICATION OF INTERMOLECULAR INTERACTIONS
US11428688B2 (en) 2018-11-07 2022-08-30 Seer, Inc. Compositions, methods and systems for protein corona analysis and uses thereof
US11965208B2 (en) * 2019-04-19 2024-04-23 Becton, Dickinson And Company Methods of associating phenotypical data and single cell sequencing data
US11906526B2 (en) 2019-08-05 2024-02-20 Seer, Inc. Systems and methods for sample preparation, data generation, and protein corona analysis
US11630112B2 (en) 2019-08-05 2023-04-18 Seer, Inc. Systems and methods for sample preparation, data generation, and protein corona analysis
US11952626B2 (en) 2021-02-23 2024-04-09 10X Genomics, Inc. Probe-based analysis of nucleic acids and proteins

Also Published As

Publication number Publication date
US11781171B1 (en) 2023-10-10
US20190093146A1 (en) 2019-03-28
US11692214B2 (en) 2023-07-04
US11512341B1 (en) 2022-11-29
US20230129372A1 (en) 2023-04-27
US11667956B2 (en) 2023-06-06
CN103703143A (zh) 2014-04-02
US20240068012A1 (en) 2024-02-29
US11926864B1 (en) 2024-03-12
EP2670863A4 (en) 2015-04-08
US20210040538A1 (en) 2021-02-11
US11708599B2 (en) 2023-07-25
US10982256B2 (en) 2021-04-20
ES2687761T3 (es) 2018-10-29
JP2014513916A (ja) 2014-06-19
US11566278B2 (en) 2023-01-31
US20230046411A1 (en) 2023-02-16
US20230051384A1 (en) 2023-02-16
US20230146787A1 (en) 2023-05-11
US11859240B2 (en) 2024-01-02
US20230313269A1 (en) 2023-10-05
US11634752B2 (en) 2023-04-25
US10626442B2 (en) 2020-04-21
CN103703143B (zh) 2016-12-14
US20230383334A1 (en) 2023-11-30
WO2012106385A3 (en) 2014-03-13
US10144950B2 (en) 2018-12-04
US11932902B2 (en) 2024-03-19
US20240068013A1 (en) 2024-02-29
US20230052354A1 (en) 2023-02-16
EP2670863B1 (en) 2018-06-27
US20200208197A1 (en) 2020-07-02
US20170321251A1 (en) 2017-11-09
JP6069224B2 (ja) 2017-02-01
US20220364151A1 (en) 2022-11-17
US20160251697A1 (en) 2016-09-01
US11732290B2 (en) 2023-08-22
US11932903B2 (en) 2024-03-19
US11939624B2 (en) 2024-03-26
US20230167484A1 (en) 2023-06-01
US20230407368A1 (en) 2023-12-21
EP2670863A2 (en) 2013-12-11

Similar Documents

Publication Publication Date Title
US11708599B2 (en) Methods of identifying multiple epitopes in cells
US20230081326A1 (en) Increasing dynamic range for identifying multiple epitopes in cells
US20220049286A1 (en) Methods of identifying multiple epitopes in cells
US20220056515A1 (en) Methods of identifying multiple epitopes in cells
US20200157603A1 (en) Methods of identifying multiple epitopes in cells
US11560585B2 (en) Methods of identifying multiple epitopes in cells

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12741485

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2013551435

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2012741485

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13981711

Country of ref document: US